Effectors of innate immunity

Abstract
The present invention provides a method of identifying agents that enhance innate immunity in a subject. The invention further provides a method of selectively supressing sepsis by suppressing expression of a proinflammatory gene while maintaining expression of an anti-inflammatory gene. Also provided are methods of identifying a polynucleotide or pattern of polynucleotides regulated by one or more sepsis or inflammatory inducing agents and inhibited by a peptide is described, methods of identifying a pattern of polynucleotide expression for inhibition of an inflammatory or septic response, and compounds and agents identified by the methods of the invention.
Description
FIELD OF THE INVENTION

The present invention relates generally to peptides and specifically to peptides effective as therapeutics and for drug discovery related to pathologies resulting from microbial infections and for modulating innate immunity or inflammation.


BACKGROUND OF THE INVENTION

Infectious diseases are the leading cause of death worldwide. According to a 1999 World Health Organization study, over 13 million people die from infectious diseases each year. Infectious diseases are the third leading cause of death in North America, accounting for 20% of deaths annually and increasing by 50% since 1980. The success of many medical and surgical treatments also hinges on the control of infectious diseases. The discovery and use of antibiotics has been one of the great achievements of modem medicine. Without antibiotics, physicians would be unable to perform complex surgery, chemotherapy or most medical interventions such as catheterization.


Current sales of antibiotics are US$26 billion worldwide. However, the overuse and sometimes unwarranted use of antibiotics have resulted in the evolution of new antibiotic-resistant strains of bacteria. Antibiotic resistance has become part of the medical landscape. Bacteria such as vancomycin-resistant Enterococcus (VRE), and methicillin-resistant Staphylococcus aureus (MRSA) strains cannot be treated with antibiotics and often, patients suffering from infections with such bacteria die. Antibiotic discovery has proven to he one of the most difficult areas for new drug development and many large pharmaceutical companies have cut back or completely halted their antibiotic development programs. However, with the dramatic rise of antibiotic resistance, including the emergence of untreatable infections, there is a clear unmet medical need for novel types of anti-microbial therapies, and agents that impact on innate immunity would be one such class of agents.


The innate immune system is a highly effective and evolved general defense system. Elements of innate immunity are always present at low levels and are activated very rapidly when stimulated. Stimulation can include interaction of bacterial signaling molecules with pattern recognition receptors on the surface of the body's cells or other mechanisms of disease. Every day, humans are exposed to tens of thousands of potential pathogenic microorganisms through the food and water we ingest, the air we breathe and the surfaces, pets and people that we touch. The innate immune system acts to prevent these pathogens from causing disease. The innate immune system differs from so-called adaptive immunity (which includes antibodies and antigen-specific B- and T-lymphocytes) because it is always present, effective immediately, and relatively non-specific for any given pathogen. The adaptive immune system requires amplification of specific recognition elements and thus takes days to weeks to respond. Even when adaptive immunity is pre-stimulated by vaccination, it may take three days or more to respond to a pathogen whereas innate immunity is immediately or rapidly (hours) available. Innate immunity involves a variety of effector functions including phagocytic cells, complement, etc, but is generally incompletely understood. Generally speaking many known innate immune responses are “triggered” by the binding of microbial signaling molecules with pattern recognition receptors such as Toll-like receptors (TLR) on the surface of host cells. We now know that Toll/Interleukin-1 Receptor (TIR) domain-containing proteins play a pivotal role in initiating aspects of the inflammatory responses. Many of these effector functions are grouped together in the inflammatory response. However, too severe an inflammatory response can result in responses that are harmful to the body, and, in an extreme case, sepsis and potentially death can occur. Thus, a therapeutic intervention to boost innate immunity, which is based on stimulation of TLR signaling (for example using a TLR agonist), has the potential disadvantage that it could stimulate a potentially harmful inflammatory response and/or exacerbate the natural inflammatory response to infection.


Early responses to infection, collectively termed innate immunity and/or acute inflammation, are substantially orchestrated by various mechanisms, for example, the interaction of bacterial molecules with TLR. It has been shown that a breakdown in the appropriate regulation of the TLR pathway can cause common chronic inflammatory diseases including inflammatory bowel disease (IBD), cardiovascular disease, arthritis, and chronic interstitial nephritis. Further, TLR engagement by conserved microbial molecules results in the translocation of the pivotal transcription factor NFκB and the transcription of ‘early-response’ genes encoding, for example, cytokines, chemokines, selected antimicrobial/host defense peptides, acute phase proteins, cell adhesion molecules, co-stimulatory molecules and proteins required for negative feedback to suppress these responses. Alternatively, an exaggerated response to bacterial stimuli underlies a clinical condition called Systemic Inflammatory Response Syndrome, or sepsis, in which high levels of cytokines and inflammatory mediators become destructive, causing organ failure, cardiovascular shock and/or death.


Sepsis occurs in approximately 780,000 patients in North America annually. Sepsis may develop as a result of infections acquired in the community such as pneumonia, or it may be a complication of the treatment of trauma, cancer or major surgery. Severe sepsis occurs when the body is overwhelmed by the inflammatory response and body organs begin to fail. Up to 120,000 deaths occur annually in the United Stated due to sepsis. Sepsis may also involve pathogenic microorganisms or toxins in the blood (e.g., septicemia), which is a leading cause of death among humans. Gram-negative bacteria are the organisms most commonly associated with such diseases. However, gram-positive bacteria are an increasing cause of infections. Gram-negative and Gram-positive bacteria and their components can all cause sepsis.


The presence of microbial components induces the release of pro-inflammatory cytokines of which tumor necrosis factor-α (TNF-α) is of extreme importance. TNF-α and other pro-inflammatory cytokines can then cause the release of other pro-inflammatory mediators and lead to an inflammatory cascade. Gram-negative sepsis is usually caused by the release of the bacterial outer membrane component, lipopolysaccharide (LPS; also referred to as endotoxin). Endotoxin in the blood, called endotoxemia comes primarily from a bacterial infection, and may be released during treatment with antibiotics. Gram-positive sepsis can be caused by the release of bacterial cell wall components such as lipoteichoic acid (LTA), peptidoglycan (PG)i rhamnose-glucose polymers made by Streptococci, or capsular polysaccharides made by Staphylococci. Bacterial or other non-mammalian DNA that, unlike mammalian DNA, frequently contains unmethylated cytosine-guanosine dimers (CpG DNA) has also been shown to induce septic conditions including the production of TNF-α. Mammalian DNA contains CpG dinucleotides at a much lower frequency, often in a methylated form. In addition to their natural release during bacterial infections, antibiotic treatment can also cause release of the bacterial cell wall components LPS and LTA and probably also bacterial DNA. This can then hinder recovery from infection or even cause sepsis.


In humans, inhalation of the Gram-negative bacterial component lipopolysaccharide (LPS), a TLR4 ligand, results in increased cytokine and chemokine (TNFα, IL1β, IL6, IL8) mRNA and protein expression within 4-6 hr of inhalation. In mutant mice lacking responsiveness to LPS animals do not develop septic shock, demonstrating that the response to endotoxin is sufficient to promote sepsis. Other TLRs exist in humans and can be engaged by other pathogen molecules to drive septic responses. For example, TLR2 is engaged by the signature cell wall-associated molecule lipoteichoic acid (LTA) from Gram positive bacteria, while DNA containing the signature dinucleotide pair unmethylated CpG engages TLR9 and can also stimulate proinflammatory Cytokine production. The nature, duration and intensity of inflammatory/septic responses are considered to involve the interplay between TLR and other receptors, different adaptor molecules such as MyD88, TIRAP/Mal and TRIF, and different signaling pathways. An ideal therapeutic regulator of the inflammatory response would be antagonistic to potentially lethal conditions such as septic shock by interacting with inflammatory signaling pathways but maintain innate immune defenses against bacterial infections, thus sustaining a balance between the protective and destructive components of inflammation.


Cationic host defense peptides (also known as antimicrobial peptides) are crucial molecules in host defense against pathogenic microbe challenge. These peptides have been demonstrated to have a wide range of functions ranging from direct antimicrobial activity to a broad range of immunomodulatory functions. They are widely distributed in nature, existing in organisms from insects to plants to mammals. The family includes defensins, cathelicidins, and histatins. Cathelicidins are small (12 to around 50 amino acids) cationic peptides and are amphipathic in nature with ˜50% hydrophobic residues. Mammalian cathelicidins are synthesized in a precursor pro-form that requires (generally-extracellular) proteolytic processing to generate the mature peptide. The only endogenous cathelicidin in humans is hCAP-18 (SEQ ID NO: 1) which is found at high concentrations in its unprocessed form (hCAP-18) in the granules of neutrophils and is processed upon degranulation and release. It is also produced by epithelial cells and keratinocytes, etc., as the hCAP-18 precursor form, and is found as the processed 37-amino acid peptide SEQ ID NO: 1 in a number of tissues and bodily fluids including gastric juices, saliva, semen, sweat, plasma, airway surface liquid and breast milk.


Cationic peptides are being increasingly recognized as a form of defense against infection, and although the major effects recognized in the scientific and patent literature were the antimicrobial effects (Hancock, R. E. W., and R. Lehrer. 1998. Cationic peptides: a new source of antibiotics. Trends in Biotechnology 16: 82-88.), it is now becoming increasingly clear that they are effectors in other aspects of innate immunity (Hancock, R. E. W. and G. Diamond. 2000. The role of cationic peptides in innate host defenses. Trends in Microbiology 8:402-410.; Hancock, R. E. W. 2001. Cationic peptides: effectors in innate immunity and novel antimicrobials. Lancet Infectious Diseases 1 :156-164).


Some cationic peptides have an affinity for binding bacterial products such as LPS and LTA. Such cationic peptides can suppress cytokine production in response to LPS, and to varying extents can prevent lethal shock. However it has not been proven as to whether such effects are due to binding of the peptides to LPS and LTA, or due to a direct interaction of the peptides with host cells. Cationic peptides are induced, in response to challenge by microbes or microbial signaling molecules like LPS, by a regulatory pathway similar to that used by the mammalian immune system (involving Toll receptors and the transcription factor; NFκB). Cationic peptides therefore appear to have a key role in innate immunity. Mutations that affect the induction of antibacterial peptides can reduce survival in response to bacterial challenge. As well, mutations of the Toll pathway of Drosophila that lead to decreased antiftingal peptide expression result in increased susceptibility to lethal fungal infections. In humans, patients with specific granule deficiency syndrome, completely lacking in α-defensins, suffer from frequent and severe bacterial infections. Other evidence includes the inducibility of some peptides by infectious agents, and the very high concentrations of such peptides that have been recorded at sites of inflammation. Cationic peptides may also regulate cell migration, to promote the ability of leukocytes to combat bacterial infections. For example, two human α-defensin peptides, HNP-1 and HNP-2, have been indicated to have direct chemotactic activity for murine and human T cells and monocytes, and human β-defensins appear to act as chemoattractants for immature dendritic cells and memory T cells through interaction with CCR6. Similarly, the porcine cationic peptide PR-39 was found to be chemotactic for neutrophils. It is unclear however as to whether peptides of different structures and compositions share these properties.


The single known cathelicidin from humans, SEQ ID NO: 1, is produced by myeloid precursors, testis, and human keratinocytes during inflammatory disorders and airway epithelium. The characteristic feature of cathelicidin peptides is a high level of sequence identity at the N-terminus prepro regions termed the cathelin domain. Cathelicidin peptides are stored as inactive propeptide precursors that, upon stimulation, are processed into active peptides.


SUMMARY OF THE INVENTION

The present invention is based on the seminal discovery that based on patterns of polynucleotide expression regulated by endotoxic lipopolysaccharide, lipoteichoic acid, CpG DNA, or other cellular components (e.g., microbe or their cellular components), and affected by cationic peptides, one can screen for novel compounds that block or reduce sepsis and/or inflammation in a subject. Further, based on the use of cationic peptides as a tool, one can identify selective enhancers of innate immunity that do not trigger the sepsis reaction and that can block/dampen inflammatory and/or septic responses.


Thus, in one embodiment, a method of identifying a polynucleotide or pattern of polynucleotides regulated by one or more sepsis or inflammatory inducing agents and inhibited by a cationic peptide, is provided. The method of the invention includes contacting cells containing polynucleotide or polynucleotides with one or more sepsis or inflammatory inducing agents and contacting the cells containing polynucleotide or polynucleotides with a cationic peptide either simultaneously or immediately thereafter. Differences in expression are detected in the presence and absence of the cationic peptide, and a change in expression, either up- or down-regulation, is indicative of a polynucleotide or pattern of polynucleotides that is regulated by a sepsis or inflammatory inducing agent and inhibited by a cationic peptide. In another aspect the invention provides a polynucleotide or polynucleotides identified by the above method. Examples of sepsis or inflammatory regulatory agents include LPS, LTA or CpG DNA or microbial components (or any combination thereof), or related agents.


In another embodiment, the invention provides a method of identi fying an agent that blocks sepsis or inflammation including combining a polynucleotide identified by the method set forth above with an agent wherein expression of the polynucleotide in the presence of the agent is modulated as compared with expression in the absence of the agent and wherein the modulation in expression affects an inflammatory or septic response.


In another embodiment, the invention provides a method of identifying a pattern of polynucleotide expression for inhibition of an inflammatory or septic response by 1) contacting cells with LPS, LTA and/or CpG DNA in the presence or absence of a cationic peptide and 2) detecting a pattern of polynucleotide expression for the cells in the presence and absence of the peptide. The pattern obtained in the presence of the peptide represents inhibition of an inflammatory or septic response. In another aspect the pattern obtained in the presence of the peptide is compared to the pattern of a test compound to identify a compound that provides a. similar pattern. In another aspect the invention provides a compound identified by the foregoing method.


In another embodiment, the invention provides a method of identifying an agent that selectively enhances innate immunity by contacting cells containing a polynucleotide or polynucleotides that encode a polypeptide involved in innate immunity, with an agent of interest, wherein expression of the polynucleotide in the presence of the agent is modulated as compared with expression of the polynucleotide in the absence of the agent and wherein the modulated expression results in enhancement of innate immunity. Preferably, the agent does not stimulate a sepsis reaction in a subject. In one aspect, the agent increases the expression of an anti-inflammatory polynucleotide. Exemplary, but non-limiting anti-inflammatory polynucleotides encode proteins such as IL-1 R antagonist homolog 1 (AI167887), IL-10 R beta (AA486393), IL-10 R alpha (U00672) TNF Receptor member 1B (AA150416), TNF receptor member 5 (H98636), TNF receptor member 11b (AA194983), IK cytokine down-regulator of HLA II (R39227), TGF-B inducible early growth response 2 (AI473938), CD2 (AA927710), IL-19 (NM013371) or IL-10 (M57627). In one aspect, the agent decreases the expression of polynucleotides encoding proteasome subunits involved in NF-κB activation such as proteasome subunit 26S (D78151). In one aspect, the agent may act as an antagonist of protein kinases. In one aspect, the agent is a peptide selected from SEQ ID NO:4-54.


In another embodiment, the invention provides a method of identifying an agent that selectively suppresses the proinflammatory response of cells containing a polynucleotide or polynucleotides that encode a polypeptide involved in innate immunity. The method includes contacting the cells with microbes, or the TLR ligands and agonists derived from those microbes, and further contacting the cells with an agent of interest, wherein the agent decreases the expression of a proinflammatory gene encoding the polynucleotide as compared with expression of the proinflammatory gene in the absence of the agent. In one aspect, the modulated expression results in suppression of proinflammatory and septic responses. Preferably, the agent does not stimulate a sepsis reaction in a subject. Exemplary, but non-limiting proinflammatory genes include TNFα, TNFAIP2, IL-1β. IL-6, NFKB1 and RELA.


In another embodiment, the invention provides a method of identifying an agent that enhances innate immunity by contacting cells containing a polynucleotide or polynucleotides that encode a polypeptide involved in innate immunity, with an agent of interest, wherein the agent suppresses inflammation and sepsis while increasing the expression of an anti-inflammatory gene encoding the polynucleotide as compared with expression of the anti-inflammatory gene in the absence of the agent and wherein the modulated expression results in enhancement of innate immunity. In one aspect, the agent inhibits the expression of proinflammatory molecules such as TNFα, IL1-β, IL-6, TNFα, TNFAIP2, or the p50 or p65 subunits of transcription factor NFκB. In another aspect, inflammation is induced by a microbe or a microbial ligand acting on a Toll-like receptor such as Toll-like receptor-2, Toll-like receptor-4, or Toll-like receptor-9. Microbial ligands include, but are not limited to a bacterial endotoxin, lipopolysaccharide, lipoteichoic acid or CpG DNA. Exemplary, but non-limiting anti-inflammatory genes include ZNF83, NFKBIA, Q9P188, INVS, DIAPHI, IER3, Q9H640, GBP2, NANS, Q86XN7, Q9H9M1, TNFAIP3, Q96MJ8, Q9BSE2, Q9H753, NTNG1, INHBE, BCL6, CXCL1, EHD1, RELB, HRK, CCL4, SESN2, NAB1, EBI3, DDX21, XBP1, SLURP1, ARS, HDAC10, MEP1A, RAP2C, GYS1, RARRES3, PPY, NFKB1, MTL4_HUMAN, Q9H040, and Q9NUP6.


In another embodiment, the invention provides a method of identifying an agent that is capable of selectively enhancing innate immunity by contacting cells containing one or more genes that encode a polypeptide involved in innate immunity and protection against an infection, with an agent of interest, wherein expression of the one or more genes in the presence of the agent is modulated as compared with expression of the one or more genes in the absence of the agent, and wherein the modulated expression results in enhancement of innate immunity. In one aspect, the invention includes agents identified by the methods. In another aspect, the agent does not stimulate a septic reaction, but does stimulate expression of the one or more genes. Exemplary, but non-limiting genes include any of the genes listed in Table 69. In one embodiment, the one or more genes encode G-coupled protein receptors that initiate signaling from extracellular ligands. Exemplary, but non-limiting genes encoding G-coupled protein receptors that initiate signaling from extracellular ligands include GPR55, GPR6, GPR30, GPCR42, CASR, and EDG2. In another embodiment, the one or more genes encode chemokines or interleukins that attract imrnune cells. Exemplary, but non-limiting genes encoding chemokines or interleukins that attract immune cells include MCP-1, MCP-3, IL-8, CXCL-1, IL-17C, and IL-19. In another embodiment, the one or more genes encode receptors for chemokines. An exemplary, but non-limiting gene encoding a receptor for chemokines includes CCR7. In another embodiment, the one or more genes encode transcription factors that mediate selective gene expression. Exemplary, but non-limiting genes encoding transcription factors that mediate selective gene expression include JAK1, STAT1, ELF1, Q9Y4C1, ETV4, POU1F1, ZNF254, ZNF292, ZNF78L1, HOXD3, and DLX5. In another embodiment, the one or more genes encode tyrosine-protein kinase or tyrosine-protein kinase receptors. Exemplary, but non-limiting genes encoding tyrosine-protein kinase or tyrosine-protein kinase receptors include MAP2K6, NTRK3, PLCG1, EFNA2, and NCK1. In another embodiment, the one or more genes encode adhesion molecules that mediate cell attachment and interaction. Exemplary, but non-limiting adhesion molecules that mediate cell attachment and interaction include the ICAM, NCAM families, and PTPRF. Exemplary, but non-limiting genes encoding adhesion molecules that mediate cell attachment and interaction include ICAM3, NCAM2, and PTPRF. In another embodiment, the one or more genes are involved in actin polymerization or cytoskeletal remodeling. Exemplary, but non-limiting genes involved in actin polymerization or cytoskeletal remodeling include Integrin-α, EPHA4, ARHGAP6, and DST. In another embodiment, the one or more genes encode regulators of transcription factors. Exemplary, but non-limiting genes encoding regulators of transcription factors include TRIP4, GMEB2, GSK3B, ARNT, BACH, ARID3A, HIPK2, POLR2D, TGIF, SSBP3, and FYB. In another embodiment, the one or more genes encode transmembrane receptors and adapters of signaling pathways. Exemplary, but non-limiting genes encoding transmembrane receptors and adapters of signaling pathways include WNT5B, FZD10, TIRAP, and REPS1. In another embodiment, the one or more genes encode proteins involved in antiviral activity. Exemplary, but non-limiting genes encoding proteins involved in antiviral activity include IFNA2, STAT1, MNDA, and IFNA2. In another embodiment, the agent stimulates the JAK-STAT pathway. In another embodiment, the agent stimulates expression of one or more genes selected from the group consisting of JAK2, STAT1, STAT3, SOCS1, and. IL-19. In another embodiment, the agent stimulates the P13K pathway. In another embodiment, the agent stimulates expression of one or more genes selected from the group consisting of BACH2/PIK3CB, Akt, CREB, IL-6, and MCP-3. In another embodiment, the agent stimulates the ERK1/2 mitogen activated kinase pathway. In another embodiment, the agent stimulates expression of one or more genes selected from the group consisting of MAP3K1 and PP2A. In another embodiment, the agent stimulates the p38 mitogen activated kinase pathway. In another embodiment, the agent stimulates expression of one or more genes selected from the group consisting of MINK1/MAP4K6, MAP2K6, and MAP2K4. In another embodiment, the agent transiently stimulates the NFκB pathway. In another embodiment, the agent stimulates expression of one or more genes selected from the group consisting of TIRAP, NFκB2 (p52), DUSP14, ICAM3, TRIP4, MMP17, ITGB4, ZNF36, ZNF251, BNIP1, CD226, NRXN1, and TNC. In another embodiment, the agent stimulates the AP-1, JNK or Wnt pathways. In another embodiment, the agent stimulates expression of one or more genes selected from the group consisting of TRIP4, TIRAP, HIPK2, GSK3B, and FZD10.


In another embodiment, the invention provides a method of identifying a pattern of gene expression for identification of an agent that selectively enhances innate immunity by contacting a cell containing one or more genes that encode a polypeptide involved in innate immunity and defense against infections, with an agent of interest, wherein expression of the one or more genes in the presence of the agent is modulated as compared with expression of the one or more genes in the absence of the agent, and wherein the modulated expression results in enhancement of innate immunity. In one embodiment, the modulated expression is a marker of enhancement of innate immunity. In another embodiment, the method further includes determining the efficacy of compounds that enhance innate immunity. In another embodiment, the one or more genes are any gene shown in Table 69. In another embodiment, the one or more genes express IL-8, IL-6, IL-19, CXCL-1, MCP-3, or MCP-1. In another embodiment, the modulated expression occurs in the presence of a bacterial signature molecule. The bacterial signature molecule may be a Toll-like receptor agonist such as bacterial lipopolysaccharide, lipoteichoic acid, and CpG bacterial signature DNA. In another embodiment, the one or more genes are any gene shown in Table 71.


In another embodiment, the invention provides a method of identifying an agent that is capable of selectively enhancing innate immunity in the presence of an infection or bacterial signature molecule by contacting a cell containing one or more genes that encode a polypeptide involved in innate immunity, with an agent of interest in the presence of a bacterial signature molecule, wherein expression of the one or more genes in the presence of the agent and bacterial signature molecule is modulated as compared with expression of the one or more genes in the absence of the agent and bacterial signature molecule, and wherein the modulated expression results in enhancement of innate immunity. In one aspect, the invention includes agents identified by the methods. In another aspect, the bacterial signature molecule is a Toll-like receptor agonist such as bacterial lipopolysaccharide, lipoteichoic acid, and CpG bacterial signature DNA. In another embodiment, the one or genes are any gene shown in Table 71. In another embodiment, the agent does not stimulate a septic reaction. In another embodiment, the agent has anti-endotoxic activity. In another embodiment, the one ore more genes are selected from the group consisting of GPD1, Q8NI35, FEZ2, NRXN1, PLCG1, Q7RTU0, ALDOB, Q9H5P1, SYT11, UBXD2, PROZ, PLAC8, Q96PN6, ASTN2, O60290, FTCD, NFKB2, CTLA4, PSMA1, CCL2, HNF4A, MAFF, FBXO32, TNFα, NPAS2, ICAM3, Q8NC30, Q81UC6, O94940, CGI-117, KDELR1, IFITM1 and COL7A1. In another embodiment, the agent stimulates transient IκBα degradation or transient NFκB subunit p50 translocation. In another embodiment, the method further includes contacting the cell with IL-1β. In another embodiment, the one or more genes encode chemokines. Exemplary, but non-limiting genes that encode chemokines include CCL20, CCL23, IL-6, and MCP-3. In another embodiment, the one or more genes encode cytokine receptors. Exemplary, but non-limiting genes that encode chemokines include EBI3 and IL7R. In another embodiment, the one or more genes encode factors involved in lymphocyte activation. Exemplary, but non-limiting genes that encode factors involved in lymphocyte activation include SLAMF1, CD58, and IL32. In another embodiment, the one or more genes encode regulators of signal transduction. Exemplary, but non-limiting genes that encode regulators of signal transduction include MAP2K2, DUSP5, MAPK8IP3, RIN2, RANBP9, IP3 3-kinase A, BATF, IRAK3, NM1, SP3, RAP2C, PNRC1, NEK1, CHC1, ZNF219, ZNF593, WIF1, PIM2, CD79A, and LATS2. In another embodiment, the one or more genes encode substrate transporters. Exemplary, but non-limiting genes that encode substrate transporters include SLC23A3 and SLC17A5. In another embodiment, the one or more genes encode apoptosis regulators. Exemplary, but non-limiting genes that encode apoptosis regulators include BOK, BIRC3, TNFRSF6, and CASP9. In another embodiment, the one or more genes encode genes associated with plasma membrane. Exemplary, but non-limiting genes that encode genes associated with plasma membrane include STIM1, BPAG1, PTPN4, TRIM36, SDK1, and FNDC5. In another embodiment, the one or more genes encode genes involved in selective ion transport and in mediating selective ion-channels. Exemplary, but non-limiting genes that encode genes involved in selective ion transport and in mediating selective ion-channels include VGCNL1, TRPC5, CACNA1B, KCNA6, KCNJ2, KCNA10, and AQP9. In another embodiment, the one or more genes encode growth modulating genes or genes involved in wound healing. xemplary, but non-limiting genes that encode growth modulating genes or genes involved in wound healing include FGF10 and AREG. In another embodiment, the one or more genes encode inflammatory mediators. Exemplary, but non-limiting genes that encode inflammatory mediators include PTGS2, SOD2, TNFAIP8, and TNIP3. In another embodiment, the method further includes contacting the cell with IL-1β, wherein the agent stimulates the PI3 kinase pathway. In another embodiment, the agent stimulates transient IκBα phosphorylation and p50 nuclear translocation. In another embodiment, the one or more genes encodes a G-protein coupled receptor or a purinergic receptor. An exemplary, but non-limiting purinergic receptor is P2X7. In another embodiment, the agent fturther stimulates phosphorylation of Akt, which stimulates activation of CREB.


In another embodiment, the invention provides a method of identifying an agent that selectively reduces inflammation by contacting a cell containing one or more genes that encode a polypeptide involved in sepsis, with an agent of interest, wherein the agent reduces expression of the one or more genes compared with expression of the one or more genes in the absence of the agent. In another embodiment, the one or more genes are selected from the group consisting of GPD1, Q8NI35, FEZ2, NRXN, PLCG1, Q7RTU0, ALDOB, Q9H5P1, SYT11, UBXD2, PROZ, PLAC8, Q96PN6, ASTN2, O60290, FTCD, NFKB2, CTLA4, PSMA1, CCL2, HNF4A, MAFF, FBXO32, TNF, NPAS2, ICAM3, Q8NC30, Q81UC6, O94940, CGI-117, KDELR1, IFITM1, and COL7A1.


In another embodiment, the invention provides a method of identifying an agent that selectively suppresses sepsis by contacting cells containing a polynucleotide or polynucleotides that encode a polypeptide involved in innate immunity, with an agent of interest, wherein the agent suppresses expression of a proinflammatory gene while maintaining expression of an anti-inflammatory gene encoding the polynucleotide as compared with expression of the anti-inflammatory gene in the absence of the agent. In one aspect, the agent inhibits the expression of proinflammatory molecules such as TNFα, IL1-β, IL-6, TNFα, TNFAIP2, or the p50 or p65 subunits of transcription factor NFκB. In another aspect, inflammation is induced by a microbe or a microbial ligand acting on a Toll-like receptor such as Toll-like receptor-2, Toll-like receptor-4, or Toll-like receptor-9. Microbial ligands include, but are not limited to a bacterial endotoxin, lipopolysaccharide, lipoteichoic acid or CpG DNA. Exemplary, but non-limiting anti-inflammatory genes include ZNF83, NFKBIA, Q9P188, INVS, DIAPH1, IER3, Q9H640, GBP2, NANS, Q86XN7, Q9H9M1, TNFAIP3, Q96MJ8, Q9BSE2, Q9H753, NTNG1, INHBE, BCL6, CXCL1, EHD1, RELB, HRK, CCL4, SESN2, NAB1, EBI3, DDX21, XBP1, SLURP1, ARS, HDAC10, MEP1A, RAP2C, GYS1, RARRES3, PPY, NFKB1, MTL4_HUMAN, Q9H040, and Q9NUP6. Exemplary, but non-limiting proinflammatory genes include LC2A6, SLC4A5, MCL1, Q86XN7, Q9H9M1, Q86UU3, Q8NAA1, C15orf2, TNFRSF5, FACL6, Q8IW99, Q96AU7, PRB4, Q9NWP0, Q8NF24, Q8TEE5, PDE4DIP, NUDT4, DUSP2, LMAN2, RELB, SNF1LK, TNFα, GHRHR, TNFSF6, ENSG00000181873, IRAK2, CKB, CASR, KRTAP4-10, ARHGEF3, CYP3A4, CYP3A7, GPR27, PAX8, GAP43, Q96M75, Q9H568, AGTRL1, C1orf22, EHD1, ADRA1B, SSTR2, SYNE1, ENSG00000139977, PTPRK, O15059, Q9NZ16, N4BP3, KIAA0341, Q8IVT2, Q9NV39, HIP1R, HIP12, KIAA0655, IL-6, TNFAIP2, RCV1, FBLN2, TWIST2, PARD6B, DCK, TULP4, LK10, SPAP1, IBRDC2, JAM2, NRG2, CBARA1, DLG2, PRKCBP1, MGLL, Q9BYE1, MARCKS, Q96N98, Q8NBY1, Q96AF2, Q9BS16, PPP2CA, RAB38, VCAM1, TTTY8, HTR2A, SERPINB10, O75121, Q9BVE1, ZCCHC2, CXCL2, GADD45B, KARS, SCG2, SLC17A2, FLT4, Q9NXT0, Q96L19, BICD1, HCK, Q8N9T8, Q9H978, PPP1R1A, PAX7, EBI3, THRA, SLC16A10, INPP5E, Q9H967, NFKB1, MKL1, SS18L2, TNFRSF9, TNFAIP6, Q9Y2K2, ING5, IL1A, TMH, HDAC4, KPTN, SEC61G, Q9Y484, FRAS1, IER5, Q8N137, Q8NCB8, Q96HQ0, Q9H5P0, TXNRD1, CAV2, SCARB1, MAP3K5, PDHX, TCEB3, C21orf55, MPHOSPH10, PDE8A, TFR2, FARP1, SERPINA1, MYO15A, RABGGTA, KCNMB4, Q9BR02, APOB, MYC, FARP2, TFAP2BL1, Q86U90, Q9H5F8, USH1C, IL-8, SOX2, Q9NVC3, NEIL2, TNIP1, ADRA1D, PCDHB9, Q12987, TNFRSF6, C20orf72, DNAJA3, MAB21 L1, BIRC2, MYST1, CNN3, CXCL3, CD80, CSRP2, RAD51L1, ADARB1, TNFSF8, Q8IW74, UXS1, ENSG00000182364, TNFRSF7, MYBL2, RAB33A, ATIC, CAMK1, CCNT1, KCNE4, BOK, NF2, PDP2, and KIAA1348.


In another embodiment, the invention provides a method of identifying an agent that selectively suppresses sepsis by contacting cells containing a polynucleotide or polynucleotides that encode a polypeptide involved in innate immunity, with an agent of interest, wherein the agent induces signaling of the JAK-STAT pathway and suppresses expression of a proinflammatory gene while maintaining expression of an anti-inflammatory gene encoding the polynucleotide as compared with expression of the anti-inflammatory gene in the absence of the agent. In one aspect, the agent inhibits the expression of proinflammatory molecules such as TNFα, NFκB2, IL1-β, IL-6, IL-8, CXCL-1, TNFAIP2, or the p50 or p65 subunits of transcription factor NFκB. In another aspect, inflammation is induced by a microbe or a microbial ligand acting on a Toll-like receptor such as Toll-like receptor-2, Toll-like receptor-4, or Toll-like receptor-9. Microbial ligands include, but are not limited to a bacterial endotoxin, lipopolysaccharide, lipoteichoic acid or CpG DNA. Exemplary, but non-limiting anti-inflammatory genes include one or more genes listed in Table 69. Exemplary, but non-limiting proinflammatory genes include one or more genes listed in Table 72.


In another embodiment, the invention provides a method of identifying a pattern of polynucleotide expression for identification of a compound that selectively enhances innate immunity. The invention includes detecting a pattern of polynucleotide expression for cells contacted in the presence and absence of a cationic peptide, wherein the pattern in the presence of the peptide represents stimulation of innate immunity; detecting a pattern of polynucleotide expression for cells contacted in the presence of a test compound, wherein a pattern with the test compound that is similar to the pattern observed in the presence of the cationic peptide, is indicative of a compound that enhances innate immunity.


In another embodiment, the invention provides a method for inferring a state of infection in a mammalian subject from a nucleic acid sample of the subject by identifying in the nucleic acid sample a polynucleotide expression pattern exemplified by an increase in polynucleotide expression of at least 2 polynucleotides in Table 50, 51 and or 52, as compared to a non-infected subject. Also included is a polynucleotide expression pattern obtained by any of the methods described above.


In another aspect a cationic peptide that is an antagonist of CXCR-4 is provided. In still another aspect, a method of identifying a cationic peptide that is an antagonist of CXCR-4 by contacting T cells with SDF-1 in the presence of absence of a test peptide and measuring chemotaxis is provided. A decrease in chemotaxis in the presence of the test peptide is indicative of a peptide that is an antagonist of CXCR-4. Cationic peptide also acts to reduce the expression of the SDF-1 receptor polynucleotide (NM012428).


In all of the above described methods, the compounds or agents of the invention include but are not limited to peptides, cationic peptides, peptidomimetics, chemical compounds, polypeptides, nucleic acid molecules and the like.


In still another aspect the invention provides an isolated cationic peptide. An isolated cationic peptide of the invention is represented by one of the following general formulas and the single letter amino acid code:

  • X1X2X3IX4PX4IPX5X2X1 (SEQ ID NO: 4), where X1 is one or two of R, L or K, X2 is one of C, S or A, X3 is one of R or P, X4 is one of A or V and X5 is one of V or W;
  • X1LX2X3KX4X2X5X3PX3X1 (SEQ ID NO: 11), where X1 is one or two of D, E, S, T or N, X2 is one or two of P, G or D, X3 is one of G, A, V, L, I or Y, X4 is one of R, K or H and X5 is one of S, T, C, M or R;
  • X1X2X3X4WX4WX4X5K (SEQ ID NO: 18), where X1 is one to four chosen from A, P or R, X2 is one or two aromatic amino acids (F, Y and W), X3 is one of P or K, X4 is one, two or none chosen from A, P, Y or W and X5 is one to three chosen from R or P;
  • X1X2X3X4X1VX3X4RGX4X3X4X1X3X1 (SEQ ID NO: 25) where X1 is one or two of R or K, X2 is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3 is C, S, M, D or A and X4 is F, I, V, M or R;
  • X1X2X3X4X1VX5X4RGX4X5X4X1X3X1 (SEQ ID NO: 32), where X1 is one or two of R or K, X2 is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3 is one of C, S, M, D or A, X4 is oneofF, I, V, M or R and X5 is one of A, I, S, M, D or R; and
  • KX1KX2FX2KMLMX2ALKKX3 (SEQ ID NO: 39), where X1 is a polar amino acid (C, S, T, M, N and Q); X2 is one of A, L, S or K and X3 is 1-17 amino acids chosen from G, A, V, L, I, P, F, S, T, K and H;
  • KWKX2X1X1X2X2X1X2X2X1X1 X2X2IFHTALKPISS (SEQ ID NO: 46), where X1 is a hydrophobic amino acid and X2 is a hydrophilic amino acid.


Additionally, in another aspect the invention provides isolated cationic peptides

KWKSFLRTFKSPVRTVFHTALKPISS(SEQ ID NO: 53)andKWKSYAHTIMSPVRLVFHTALKPISS.(SEQ ID NO: 54)


Also provided are nucleic acid sequences encoding the cationic peptides of the invention, vectors including such polynucleotides and host cells containing the vectors.


In another embodiment, the invention provides methods for stimulating or enhancing innate immunity in a subject comprising administering to the subject a peptide of the invention, for example, peptides set forth in SEQ ID NO:1-4, 11, 18, 25, 32, 39, 46, 53 or 54. As shown in the Examples herein, innate immunity can be evidenced by monocyte activation, proliferation, differentiation, or MAP kinase pathway activation just by way of example. In one aspect, the method includes further administering a serum factor such as GM-CSF to the subject. The subject is preferably any mammal and more particularly a human subject.


In another embodiment, the invention provides a method of stimulating innate immunity in a subject having or at risk of having an infection including administering to the subject a sub-optimal concentration of an antibiotic in combination with a peptide of the invention. In one aspect, the peptide is SEQ ID NO:1 or SEQ ID NO:7.


In all of the above described embodiments, the methods may be performed ex vivo.




BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 demonstrates the synergy of SEQ ID NO: 7 with cefepime in curing S. aureus infections. CD-1 mice (8/group) were given 1×107 S. aureus in 5% porcine mucin via IP injection. Test compound (50 μg-2.5 mg/kg) was given via a separate IP injection 6 hours after S. aureus. At this time Cefepime was also given at a dose of 0.1 mg/kg. Mice were euthanized 24 hr later, blood removed and plated for viable counts. The average±standard error is shown. This experiment was repeated twice.



FIG. 2 shows exposure to SEQ ID NO: 1 induces phosphorylation of ERK1/2 and p38. Lysates from human peripheral blood derived monocytes were exposed to 50 μg/ml of SEQ ID NO: 1 for 15 minutes. A) Antibodies specific for the phosphorylated forms of ERK and p38 were used to detect activation of ERK1/2 and p38. All donors tested showed increased phosphorylation of ERK1/2 and p38 in response to SEQ ID NO: 1 treatment. One representative donor of eight is shown. Relative amounts of phosphorylation of ERK (B) and p38(C) were determined by dividing the intensities of the phosphorylated bands by the intensity of the corresponding control band as described in the Materials and Methods in Example 12.



FIG. 3 shows SEQ ID NO: 1 induced phosphorylation of ERK1/2 does not occur in the absence of serum and the magnitude of phosphorylation is dependent upon the type of serum present. Human blood derived monocytes were treated with 50 μg/ml of SEQ ID NO: 1 for 15 minutes. Lysates were run on a 12% acrylamide gel then transferred to nitrocellulose membrane and probed with antibodies specific for the phosphorylated (active) form of the kinase. To normalize for protein loading, the blots were reprobed with β-actin. Quantification was done with ImageJ software. The FIG. 3 insert demonstrates that SEQ ID NO: 1 is unable to induce MAPK activation in human monocytes under serum free conditions. Cells were exposed to 50 mg/ml of SEQ ID NO: 1 (+), or endotoxin free water (−) as a vehicle control, for 15 minutes. (A) After exposure to SEQ ID NO: 1 in media containing 10% fetal calf serum, phosphorylated ERK1/2 was detectable, however, no phosphorylation of ERK1/2 was detected in the absence of serum (n=3). (B) Elk-1, a transcription factor downstream of ERK1/2, was activated (phosphorylated) upon exposure to 50 μg/ml of SEQ ID NO: 1 in media containing 10% fetal calf serum, but not in the absence of serum (n=2).



FIG. 4 shows SEQ ID NO: 1 induced activation of ERK1/2 occurs at lower concentrations and is amplified in the presence of certain cytokines. When freshly isolated monocytes were stimulated in media containing both GM-CSF (100 ng/ml) and IL-4 (100 ng/ml) SEQ ID NO: 1 induced phosphorylation of ERK1/2 was apparent at concentrations as low as 5 μg/ml. This synergistic activation of ERK1/2 seems to be due primarily to GM-CSF.



FIG. 5 shows peptide affects both transcription of various cytokine genes and release of IL-8 in the 16HBE4o-human bronchial epithelial cell line. Cells were grown to confluency on a semi-permeable membrane and stimulated on the apical surface with 50 μg/ml of SEQ ID NO: 1 for four hours. A) SEQ ID NO: 1 treated cells produced significantly more IL-8 than controls, as detected by ELISA in the supernatant collected from the apical surface, but not from the basolateral surface. Mean±SE of three independent experiments shown, asterisk indicates p=0.002. B) RNA was collected from the above experiments and RT-PCR was performed. A number of cytokine genes known to be regulated by either ERK1/2 or p38 were up-regulated upon stimulation with peptide. The average of two independent experiments is shown.



FIG. 6 is a graphical representation showing that SEQ ID NO: 1 suppresses LPS-induced secretion of TNF-α. The concentration of the pro-inflammatory cytokine TNFα (Y-axis) was monitored in the tissue culture supernatant or cytoplasmic extracts of cells by ELISA. The results are an average (±standard deviation) of three independent experiments. (A) THP-1 cells were stimulated with 10 ng/ml (-●-) or 100 ng/ml (-▪-) of LPS in the presence of increasing concentrations of SEQ ID NO: 1 (X-axis) for 4 hr. (B) PBMCs were stimulated with 100 ng/ml of LPS in presence or absence of 20 μg/ml SEQ ID NO: 1 for 4 hrs. The anti-endotoxin effect of SEQ ID NO: 1 demonstrated in PBMC was statistically significant with p-value of <0.05 (**). (C) THP-1 cells were treated with LPS, SEQ ID NO: 1 or LPS+ SEQ ID NO: 1 for 4 hr in the absence (white bar) or presence of actinomycin D (black bar), the effect of actinomycin D on LPS-induced TNFα secretion was statistical significant with p-value<0.001 (***). (D) Cytoplasmic extracts of THP-1 cells treated with LPS, SEQ ID NO: 1 or LPS+ SEQ ID NO: 1 for 60 mins in the absence (black bar) or presence of monensin (white bar) were monitored by ELISA.



FIG. 7 is a graphical representation showing the anti-endotoxic effect of SEQ ID NO: 1 involves pre- and post-transcriptional events. Tissue culture supernatants were screened for TNFα by ELISA following stimulation of cells with 100 ng/ml of LPS in the absence (-▪-) or in the presence of 20 μg/ml SEQ ID NO: 1 (-●-) for 1, 2, 4 and 24 hr of treatment. In each case, the control indicates un-stimulated cells (-▾-), the y-axis represents TNFα concentration and the x-axis indicates time (hr). SEQ ID NO: 1 (20 ug/ml) was added (A) simultaneously with LPS, (B) after 30 min of LPS treatment, or (C) 30 min prior to LPS treatment. See materials and method for details. The results are an average (±standard deviation) of 3 independent experiments.



FIG. 8 is a graphical representation showing that SEQ ID NO: 1 modifies inflammatory agent-induced cytokine secretion by PBMC. PBMC were incubated alone or with TLR agonists (LPS, LTA, CpG) or inflammatory cytokines (TNFα, IL1β) for 4 or 24 hr in the presence (black bars) or absence (white bars) of SEQ ID NO: 1. See materials and methods in Example 13 for details. The concentration (y-axis) of IL1α, IL6, IL8 and TNFα(x-axis) were measured in the tissue culture supernatants by multiplex bead ELISA. The results are an average (±standard deviation) of 3 independent experiments. The effect of SEQ ID NO: 1 on agonist induced cytokine production was statistical significant with p-value<0.05 (***), p<0.1 (**) or p<0.15 (*).



FIG. 9 is a graphical representation showing an LPS-induced gene transcription profile in monocytes is altered by the presence of host defense peptide SEQ ID NO: 1. (A) THP-1 cells were stimulated with 100 ng/ml LPS in the absence (top panel) or presence (lower panel) of 20 ug/ml SEQ ID NO: 1 for 1, 2, 4 or 24 hr. Using microarray analysis, the gene expression in response to stimuli was calculated relative to that in unstimulated cells at each time point. The relative gene expression is overlaid on the TLR-4 protein network using the supervised clustering tool Cytoscape. The colour code for the fold change and identification of proteins are in the left panel. (B) Cluster analysis of the differentially expressed genes as measured using log ratio (y-axis) of microarray spot intensity, with NFκB binding sites in response to 100 ng/ml of LPS in the absence (top) or presence of 20 ug/ml of SEQ ID NO: 1 (bottom) based on similar temporal expression profiles over the time course of I to 24 hr (x-axis) using K-means, a no-hierarchical algorithm with an affinity threshold of 85%. The table indicates the total number of differentially expressed genes, total number of clusters, number of clusters containing genes with NFκB binding sites and the NFκB target genes found in the clusters.



FIG. 10 is a graphical representation showing that SEQ ID NO: 1 selectively modulates the transcription of LPS-induced pro-inflammatory genes. qPCR of gene expression in LPS-stimulated cells (-▪-), cells treated with SEQ ID NO: 1 alone (-▾-) or cells treated with a combination of LPS and SEQ ID NO: 1 (-●-) for 1,2,4, and 24 hr (x-axis). Results shown are an average (±standard error) of three independent experiments. Fold changes (y-axis, log scale) for each gene were normalized to GAPDH and are relative to the gene expression in un-stimulated cells (normalized to 1) using the comparative Ct method (see materials and methods in Example 13 for details).



FIG. 11 is a pictorial diagram and a graphical representation showing that SEQ ID NO: 1 suppresses LPS-induced translocation of NFκB subunits p50 and p65. (A) Western blot of NFκB subunits (identified on the right) in the nuclear extract of THP-1 cells following incubation in the absence (−) or presence (+) of 100 ng/ml LPS or LPS and 20 μg/ml SEQ ID NO: 1 for 60 mins. Pre-stained molecular mass markers are indicated on the left. (B) ELISA for NFκB subunit p50 (upper panel) and NFκB subunit p65 (lower panel) detected in the nuclear extracts of THP-1 cells stimulated for 60 min as described in (A). The y-axis represents relative light units (luminescence). See materials and methods in Example 13 for details. Results are representative of 3 independent experiments.



FIG. 12 is a pictorial diagram of a model describing mechanisms of anti-endotoxin activity of SEQ ID NO: 1. Based on the data presented herein, SEQ ID NO: 1 regulates LPS-induced gene transcription and cytokine production, by one or more of several mechanisms. (1) SEQ ID NO: 1 can interact directly with LPS to reduce its binding to LBP, MD2 or another component of the TLR4 receptor complex, thus reducing activation of the downstream pathway. (2) SEQ ID NO: 1 partially inhibits the TLR4→NFκB pathway and LPS-induced p50/p65 translocation probably by the action of certain negative regulators of NFκB (TNFAIP3, NFKBIA), the expression of which is relatively unaffected by SEQ ID NO: 1. (3) SEQ ID NO: 1 selectively modulates gene transcription; completely inhibiting certain pro-inflammatory genes (NFKB-1 (p50), TNFAIP2) and reducing the expression of others (TNFα). (4) SEQ ID NO: 1 directly triggers MAP kinase pathways that can impact on pro-inflammatory pathways. (5) SEQ ID NO: 1 has a stronger effect on e.g. TNFα protein production than on TNFα gene expression, and thus may directly or indirectly influence protein translation, stabilization, or processing. Points of intervention by SEQ ID NO: 1 are indicated by activation custom characterinhibition (⊥), or suppression (→). Other abbreviations used are phosphorylation (P) and ubiquitination (custom character).



FIG. 13 is a pictorial diagram of a model describing mechanisms in which host defense peptides induce gene expression of the Janus Kinases and STAT family of transcription factors. Human PBMC were stimulated with (I) the human host defense peptide LL-37 (20 μg/ml) and (2) peptide SEQ ID NO: 7 for 4 hr. Using microarray analysis, the gene expression in CD14+ monocytes purified from the PBMC population in response to stimuli was calculated relative to that in un-stimulated cells. Differentially expressed genes were those with a fold change over the untreated control of 1.5-fold and a p-value<0.06 (calculated using a two-sided one-sample Student t-test on the log2-ratios within each treatment group). The relative gene expression was overlaid onto a protein network using the systems biology clustering software tool Metacore™ (GeneGo, Inc., CA, USA). The color code for the fold changes are indicated as up-regulation (red) and down-regulation (blue) in response to the stimuli.



FIG. 14 is a graphical representation showing that SEQ ID NO: 7 induces transcription of genes functional in immune response. Quantitative real-time PCR of gene expression in human CDI4+ monocytes in response to host defense peptide SEQ ID NO: 7 after 4 hr of stimulation. Results shown are from four independent biological replicates (X-axis). Fold changes (Y-axis) for each gene were normalized to GAPDH and are relative to the gene expression in un-stimulated cells (normalized to 1) using the comparative Ct method. These represent markers of SEQ ID NO: 7 effects on blood cells.



FIG. 15 is a graphical representation showing that SEQ ID NO: 7 induces protein production in human PBMC within 4 hr of stimulation. PBMC were stimulated with SEQ ID NO: 7 (200 μg/ml) for 4 hours. The concentration (Y-axis) of cytokines IL-6 and IL-8 were measured in tissue culture supernatants by ELISA from PBMC of four individual donors (X-axis). The results shown are from four independent experiments.



FIG. 16 is a graphical representation showing that LPS-induced transcriptional responses in human monocytes are suppressed in the presence of SEQ ID NO: 7. Quantitative real-time PCR of gene expression in human CD14+ monocytes in response to LPS in the presence and absence of host defense peptide of SEQ ID NO: 7 after 4 hr of stimulation. Results shown are from four independent biological replicates (X-axis). Fold changes (Y-axis) for each gene were normalized to GAPDH, and are relative to the gene expression in un-stimulated cells (normalized to 1) using the comparative Ct method.



FIGS. 17A and 17B are graphical representations showing that SEQ ID NO: 7 suppresses LPS-induced pro-inflammatory TNF-o: secretion in human mononuclear cells within 4 hours of stimulation. Human PBMC and Human monocytic THP-1 cells were stimulated with LPS in the presence and absence of SEQ ID NO: 7 for 4 hours. The cells were treated with the peptide 45 mins prior to LPS stimulation. The concentration (Y-axis) of cytokines TNF-α was measured in tissue culture supernatants by ELISA. The results shown are from PBMC of three independent human donors. The results are an average (±standard deviation) of three independent experiments in THP-1 cells.



FIG. 18 is a graphical representation of a Venn diagram showing that the human host defense peptide LL-37 demonstrates both overlapping and distinct induction of differentially expressed (DE) and statistically significant genes compared to SEQ ID NO: 7.



FIG. 19 is a pictorial diagram showing that protein levels of total IιBα diminish within 30 min and return to control levels by 60 min in THP-1 cells, indicating that LL-37 may directly modulate elements of the LPS signaling pathway.



FIGS. 20A and 20B are graphical representations showing data from fresh isolated human PBMCs that were incubated with IL-1β (10 ng/ml) or LPS (100 ng/ml) in absence or presence of LL-37 (20 ug/ml) for 24 hours. IL-6 and MCP-3 ELISA were performed to measure the level of protein release.



FIGS. 21A and 21B are pictorial diagrams showing Western blots for cytoplasm protein and nuclear protein. The combined treatment of LL-37 and IL-1 β showed higher IκBα phosphorylation after 30 min and p50 nuclear translocation after 60 min than LL-37 or IL-1β treatment alone in human PBMCs. Similar translocation results were also observed in THP-1 cells at an earlier time point (20 min after treatment). In addition, LL-37 alone induced NFκB subunit p50 translocation in both human PBMCs and THP-1 cells.



FIGS. 22A-22D are graphical representations showing data from human PBMCs that were pre-treated with PI3 kinase inhibitor, LY294002 (25 μM) for 1 h, followed by incubation with IL-1β (10 ng/ml) in the presence or absence of LL-37 (20 μg/ml) for 24 hours. The LL-37 plus IL-1β mediated production of IL-6 and MCP-3 was significantly inhibited by LY294002 pre-incubation, indicating that P13 kinase plays a role in LL-37-induced modulation of cytokine and chemokine production.



FIG. 23 is a pictorial diagram showing that activation of PI3 kinase causes activation of a number of intracellular signal transduction pathways, including phosphorylation of the downstream target protein kinase B (Akt). Further analysis showed that phosphorylation of CREB was observed in human PBMCs after exposure to LL-37 for 30min and 60min. Activation of Akt and CREB was augmented by the presence of IL-1β (10 ng/ml).




DETAILED DESCRIPTION OF THE INVENTION

The present invention provides novel cationic peptides, characterized by a group of generic formulas (SEQ ID NO: 4, 11, 18, 25, 32, 39, 46), which have ability to modulate (e.g., up- and/or down regulate) polynucleotide expression, thereby regulating sepsis and inflammatory responses and/or innate immunity.


“Innate immunity” as used herein refers to the natural ability of an organism to defend itself against invasions by pathogens. Pathogens or microbes as used herein, may include, but are not limited to bacteria, fungi, parasite, and viruses. Innate immunity is contrasted with acquired/adaptive immunity in which the organism develops a defensive mechanism based substantially on antibodies and/or immune lymphocytes that is characterized by specificity, amplifiability and self vs. non-self dsicrimination. With innate immunity, broad, nonspecific immunity is provided and there is no immunologic memory of prior exposure. The hallmarks of innate immunity are effectiveness against a broad variety of potential pathogens, independence of prior exposure to a pathogen, and immediate effectiveness (in contrast to the specific immune response which takes days to weeks to be elicited). In addition, innate immunity includes immune responses that affect other diseases, such as cancer, inflammatory diseases, multiple sclerosis, various viral infections, and the like.


As used herein, the term “cationic peptide” refers to a sequence of amino acids from about 5 to about 50 amino acids in length. In one aspect, the cationic peptide of the invention is from about 10 to about 35 amino acids in length. A peptide is “cationic” if it possesses sufficient positively charged amino acids to have a pI greater than about 9.0, where pl (isoelectric point)=pH when the net charge of the peptide is neutral. Typically, at least two of the amino acid residues of the cationic peptide will be positively charged, for example, lysine or arginine. “Positively charged” refers to the side chains of the amino acid residues which have a net positive charge at pH 7.0. Examples of naturally occurring cationic antimicrobial peptides which can be recombinantly produced according to the invention include defensins, cathelicidins, magainins, melittin, and cecropins, bactenecins, indolicidins, polyphemusins, tachyplesins, and analogs thereof. A variety of organisms make cationic peptides, molecules used as part of a non-specific defense mechanism against microorganisms. When isolated, these peptides are toxic to a wide variety of microorganisms, including bacteria, fuingi, and certain enveloped viruses. While cationic peptides act against many.pathogens, notable exceptions and varying degrees of toxicity exist. However this patent reveals additional cationic peptides with no toxicity towards microorganisms but an ability to protect against infections through stimulation of innate immunity, and this invention is not limited to cationic peptides with antimicrobial activity. In fact, many peptides useful in the present invention do not have antimicrobial activity.


Cationic peptides known in the art include for example, the human cathelicidin LL-37, and the bovine neutrophil peptide indolicidin and the bovine variant of bactenecin, Bac2A.

(SEQ ID NO: 1)LL-37LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES(SEQ ID NO: 2)IndolicidinILPWKWPWWPWRR-NH2(SEQ ID NO: 3)Bac2ARLARIVVIRVAR-NH2


Although SEQ ID NO: 1 is often defined as an antimicrobial (direct killing) peptide it has been suggested that at physiological salt conditions, this peptide is not antimicrobial at the concentrations (1-5 μg/ml) normally found in adults at mucosal surfaces (Bowdish, D. M. E., D. J. Davidson, Y. E. Lau, K. Lee, M. G. Scott, and R. E. W. Hancock. 2005. Impact of LL-37 on anti-infective immunity. J. Leukocyte Biol. 77:451-459). Moreover under these conditions and at these concentrations, SEQ ID NO: 1 exhibits a variety of immunomodulatory functions. This could help to explain why SEQ ID NO: 1 administration can protect mice against certain bacterial infections, due to its ability to modulate immunity. SEQ ID NO: 1 is also able to protect mice and rats against endotoxemialsepsis induced by pure LPS indicating that SEQ ID NO: 1 can suppress potentially harmful pro-inflammatory responses.


Accordingly, the present invention provides evidence that human host defense peptide SEQ ID NO: 1 has potent anti-endotoxin properties, at very low (≦1 μg/ml) concentrations and physiological salt conditions reflecting those found in vivo. It is further demonstrated here that SEQ ID NO: 1 had a general anti-inflammatory effect on TLR stimulation, inhibiting pro-inflammatory cytokine release from human monocytic cells stimulated with TLR2, TLR4 and TLR9 agonists. The suppression of inflammatory responses by SEQ ID NO: 1 in LPS-stimulated cells is selective, as SEQ ID NO: 1 does not block the expression of certain (pro-inflammatory) genes required for cell recruitment and movement, yet abrogates pro-inflammatory cytokine responses that can potentially lead to sepsis. The anti-inflammatory activity of SEQ ID NO: 1 is apparently mediated through a diversity of mechanisms.


In innate immunity, the immune response is not dependent upon antigens. The innate immunity process may include the production of secretory molecules and cellular components as set forth above. In innate immunity, the pathogens are recognized by receptors (for example, Toll-like receptors) that have broad specificity, are capable of recognizing many pathogens, and are encoded in the germline. These Toll-like receptors have broad specificity and are capable of recognizing many pathogens. When cationic peptides are present in the immune response, they aid in the host response to pathogens. This change in the immune response induces the release of chemokines, which promote the recruitment of immune cells to the site of infection.


Chemokines, or chemoattractant cytokines, are a subgroup of immune factors that mediate chemotactic and other pro-inflammatory phenomena (See, Schall, 1991, Cytokine 3:165-183). Chemokines are small molecules of approximately 70-80 residues in length and can generally be divided into two subgroups, α which have two N-terminal cysteines separated by a single amino acid (CxC) and β which have two adjacent cysteines at the N terminus (CC). RANTES, MIP-1α and MIP-1β are members of the β subgroup (reviewed by Horuk, R., 1994, Trends Pharmacol. Sci, 15:159-165; Murphy, P. M., 1994, Annu. Rev. Immunol., 12:593-633). The amino terminus of the β chemokines RANTES, MCP-1, and MCP-3 have been implicated in the mediation of cell migration and inflammation induced by these chemokines. This involvement is suggested by the observation that the deletion of the amino terminal 8 residues of MCP-1, amino terminal 9 residues of MCP-3, and amino terminal 8 residues of RANTES and the addition of a methionine to the amino terminus of RANTES, antagonize the chemotaxis, calcium mobilization and/or enzyme release stimulated by their native counterparts (Gong et al., 1996 J. Biol. Chem. 271:10521-10527; Proudfoot et al., 1996 J Biol. Chem. 271:2599-2603). Additionally, α chemokine-like chemotactic activity has been introduced into MCP-1 via a double mutation of Tyr 28 and Arg 30 to leucine and valine, respectively, indicating that internal regions of this protein also play a role in regulating chemotactic activity (Beall et al., 1992, J. Biol. Chem. 267:3455-3459).


The monomeric forms of all chemokines characterized thus far share significant structural homology, although the quaternary structures of α and β groups are distinct. While the monomeric structures of the β and a chemokines are very similar, the dimeric structures of the two groups are completely different. An additional chemokine, lymphotactin, which has only one N terminal cysteine has also been identified and may represent an additional subgroup (γ) of chemokines (Yoshida et al., 1995, FEBS Lett. 360:155-159; and Kelner et al., 1994, Science 266:1395-1399).


Receptors for chemokines belong to the large family of G-protein coupled, 7 transmembrane domain receptors (GCR's) (See, reviews by Horuk, R., 1994, Trends Pharmacol. Sci. 15:159-165; and Murphy, P. M., 1994, Annu. Rev. Immunol. 12:593-633). Competition binding and cross-desensitization studies have shown that chemokine receptors exhibit considerable promiscuity in ligand binding. Examples demonstrating the promiscuity among β chemokine receptors include: CC CKR-1, which binds RANTES and MIP-1α (Neote et al., 1993, Cell 72: 415-425), CC CKR-4, which binds RANTES, MIP-1α, and MCP-1 (Power et al., 1995, J. BioL. Chem. 270:19495-19500), and CC CKR-5, which binds RANTES, MIP-1α, and MIP-1β (Alkhatib et al., 1996, Science, in press and Dragic et al., 1996, Nature 381:667-674). Erythrocytes possess a receptor (known as the Duffy antigen) which binds both α and β chemokines (Horuk et al., 1994, J. Biol. Chem. 269:17730-17733; Neote et al., 1994, Blood 84:44-52; and Neote et al., 1993, J. Biol. Chem. 268:12247-12249). Thus the sequence and structural homologies evident among chemokines and their receptors allows some overlap in receptor-ligand interactions.


In one aspect, the present invention provides the use of compounds including peptides of the invention to reduce sepsis and inflammatory responses by acting directly on host cells. In this aspect, a method of identification of a polynucleotide or polynucleotides that are regulated by one or more sepsis or inflammatory inducing agents is provided, where the regulation is altered by a cationic peptide. Such sepsis or inflammatory inducing agents include, but are not limited to endotoxic lipopolysaccharide (LPS), lipoteichoic acid (LTA) and/or CpG DNA or intact bacteria or other bacterial components. The identification is performed by contacting the polynucleotide or polynucleotides with the sepsis or inflammatory inducing agents and further contacting with a cationic peptide either simultaneously or immediately after. The expression of the polynucleotide in the presence and absence of the cationic peptide is observed and a change in expression is indicative of a polynucleotide or pattern of polynucleotides that is regulated by a sepsis or inflammatory inducing agent and inhibited by a cationic peptide. In another aspect, the invention provides a polynucleotide identified by the method.


Once identified, such polynucleotides will be useful in methods of screening for compounds that can block sepsis or inflammation by affecting the expression of the polynucleotide. Such an effect on expression may be either up regulation or down regulation of expression. By identifying compounds that do not trigger the sepsis reaction and that can block or dampen inflammatory or septic responses, the present invention also presents a method of identifying enhancers of innate immunity. Additionally, the present invention provides compounds that are used or identified in the above methods.


Candidate compounds are obtained from a wide variety of sources including libraries of synthetic. or natural compounds. For example, numerous means are available for, random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, and the like to produce structural analogs. Candidate agents are also found among biomolecules including, but not limited to: peptides, peptidiomimetics, saccharides, fatty acids, steroids, purines, pyrimidines, polypeptides, polynucleotides, chemical compounds, derivatives, structural analogs or combinations thereof.


Incubating components of a screening assay includes conditions which allow contact between the test compound and the polynucleotides of interest. Contacting includes in solution and in solid phase, in a cell, or on a cell surface. The test compound may optionally be a combinatorial library for screening a plurality of compounds. Compounds identified in the method of the invention can be further evaluated, detected, cloned, sequenced, and the like, either in solution or after binding to a solid support, by any method usually applied to the detection of a compound.


Generally, in the methods of the invention, a cationic peptide is utilized to detect and locate a polynucleotide that is essential in the process of sepsis or inflammation. Once identified, a pattern of polynucleotide expression may be obtained by observing the expression in the presence and absence of the cationic peptide. The pattern obtained in the presence of the cationic peptide is then useful in identifying additional compounds that can inhibit expression of the polynucleotide and therefore block sepsis or inflammation. It is well known to one of skill in the art that non-peptidic chemicals and peptidomimetics can mimic the ability of peptides to bind to receptors and enzyme binding sites and thus can be used to block or stimulate biological reactions. Where an additional compound of interest provides a pattern of polynucleotide expression similar to that of the expression in the presence of a cationic peptide, that compound is also useful in the modulation of sepsis or an innate immune response. In this manner, the cationic peptides of the invention, which are known inhibitors of sepsis and inflammation and enhancers of innate immunity are useful as tools in the identification of additional compounds that inhibit sepsis and inflammation and enhance innate immunity.


As can be seen in the Examples below, peptides of the invention have a widespread ability to reduce the expression of polynucleotides regulated by LPS. High levels of endotoxin in the blood-are responsible for many of the symptoms seen during a serious infection or inflammation such as fever and an elevated white blood cell count. Endotoxin is a component of the cell wall of Gram-negative bacteria and is a potent trigger of the pathophysiology of sepsis. The basic mechanisms of inflammation and sepsis are related. In Example 1, polynucleotide arrays were utilized to determine the effect of cationic peptides on the transcriptional response of epithelial cells. Specifically, the effects on over 14,000 different specific polynucleotide probes induced by LPS were observed. The tables show the changes seen with cells treated with peptide compared to control cells. The resulting data indicated that the peptides have the ability to reduce the expression of polynucleotides induced by LPS.


Example 2, similarly, shows that peptides of the invention are capable of neutralizing the stimulation of immune cells by Gram positive and Gram negative bacterial products. Additionally, it is noted that certain pro-inflammatory polynucleotides are down-regulated by cationic peptides, as set forth in table 24 such as TLR1 (AI339155), TLR2 (T57791), TLR5 (N41021), TNF receptor-associated factor 2 (T55353), TNF receptor-associated factor 3 (AA504259), TNF receptor superfamily, member 12 (W71984), TNF receptor superfamily, member 17 (AA987627), small inducible cytokine subfamily B, member 6 (AI889554), IL-12R beta 2 (AA977194), IL-18 receptor 1 (AA482489), while anti-inflammatory polynucleotides are up-regulated by cationic peptides, as seen in table 25 such as IL-1 R antagonist homolog 1 (AI167887), IL-10 R beta (AA486393), TNF Receptor member 1B (AA150416), TNF receptor member 5 (H98636), TNF receptor member 11b (AA194983), IK cytokine down-regulator of HLA II (R39227), TGF-B inducible early growth response 2 (AI473938), or CD2 (AA927710). The relevance and application of these results are confirmed by an in vivo application to mice.


In another aspect, the invention provides a method of identifying an agent that enhances innate immunity. In-the method, a polynucleotide or polynucleotides that encode a polypeptide involved in innate immunity is contacted with an agent of interest. Expression of the polynucleotide is determined, both in the presence and absence of the agent. The expression is compared and of the specific modulation of expression was indicative of an enhancement of innate immunity. In another aspect, the agent does not stimulate a septic reaction as revealed by the lack of upregulation of the pro-inflammatory cytokine TNF-α. In still another aspect the agent reduces or blocks the inflammatory or septic response. In yet another aspect, the agent reduces the expression of TNF-αand/or interleukins including, but not limited to, IL-1β, IL-6, IL-12 p40, IL-12 p70, and IL-8.


In another aspect, the invention provides methods of direct polynucleotide regulation by cationic peptides and the use of compounds including cationic peptides to stimulate elements of innate immunity. In this aspect, the invention provides a method of identification of a pattern of polynucleotide expression for identification of a compound that enhances innate immunity. In the method of the invention, an initial detection of a pattern of polynucleotide expression for cells contacted in the presence and absence of a cationic peptide is made. The pattern resulting from polynucleotide expression in the presence of the peptide represents stimulation of innate immunity. A pattern of polynucleotide expression is then detected in the presence of a test compound, where a resulting pattern with the test compound that is similar to the pattern observed in the presence of the cationic peptide is indicative of a compound that enhances innate immunity. In another aspect, the invention provides compounds that are identified in the above methods. In another aspect, the compound of the invention stimulates chemokine or chemokine receptor expression. Chemokine or chemokine receptors may include, but are not limited to CXCR4, CXCR1, CXCR2, CCR2, CCR4, CCR5, CCR6, MIP-1 alpha, MDC, MIP-3 alpha, MCP-1, MCP-2, MCP-3, MCP4, MCP-5, anid RANTES. In still another aspect, the compound is a peptide, peptidomimetic, chemical compound, or a nucleic acid molecule.


In still another aspect the polynucleotide expression pattern includes expression of pro-inflammatory polynucleotides. Such pro-inflammatory polynucleotides may include, but are not limited to, ring finger protein 10 (D87451), serine/threonine protein kinase MASK (AB040057), KIAA0912 protein (AB020719), KIAA0239 protein (D87076), RAP1, GTPase activating protein 1 (M64788), FEM-1-like death receptor binding protein (AB007856), cathepsin S (M90696), hypothetical protein FLJ20308 (AK000315), pim-1 oncogene (M54915), proteasome subunit beta type 5 (D2901 1), KIAA0239 protein (D87076), mucin 5 subtype B tracheobronchial (AJ001403), cAMP response element-binding protein CREBPa, integrin alpha M (J03925), Rho-associated kinase 2 (NM004850), PTD017 protein (AL050361) unknown genes (AK00143, AK034348, AL049250, AL161991, AL031983) and any combination thereof In still another aspect the polynucleotide expression pattern includes expression of cell surface receptors that may include but is not limited to retinoic acid receptor (X06614), G protein-coupled receptors (Z94155, X81892, U52219, U22491, AF015257, U66579) chemokine (C-C motif) receptor 7 (L31584), tumor necrosis factor receptor superfamily member 17 (Z29575), interferon gamma receptor 2 (U05875), cytokine receptor-like factor 1 (AF059293), class I cytokine receptor (AF053004), coagulation factor II (thrombin) receptor-like 2 (U9297 1), leukemia inhibitory factor receptor (NM002310), interferon gamma receptor 1 (AL050337).


In Example 4 it can be seen that the cationic peptides of the invention alter polynucleotide expression in macrophage and epithelial cells. The results of this example-show that pro-inflammatory polynucleotides are down-regulated by cationic peptides (Table 24) whereas anti-inflammatory polynucleotides are up-regulated by cationic peptides (Table 25).


It is shown below, for example, in tables 1-15, that cationic peptides can neutralize the host response to the signaling molecules of infectious agents as well as modify the transcriptional responses of host cells, mainly by down-regulating the pro-inflammatory response and/or up-regulating the anti-inflammatory response. Example 5 shows that the cationic peptides can aid in the host response to pathogens by inducing the release of chemokines, which promote the recruitment of immune cells to the site of infection. The results are confirmed by an in vivo application to mice.


It is seen from the examples below that cationic peptides have a substantial influence on the host response to pathogens in that they assist in regulation of the host immune response by inducing selective pro-inflammatory responses that for example promote the recruitment of immune cells to the site of infection but not inducing potentially harmful pro-inflammatory cytokines. Sepsis appears to be caused in part by an overwhelming pro-inflammatory response to infectious agents. Peptides can aid the host in a “balanced” response to pathogens by inducing an anti-inflammatory response and suppressing certain potentially harmful pro-inflammatory responses.


In Example 7, the activation of selected MAP kinases was examined, to study the basic mechanisms behind the effects of interaction of cationic peptides with cells. Macrophages activate MEK/ERK kinases in response to bacterial infection. MEK is a MAP kinase kinase that when activated, phosphorylates the downstream kinase ERK (extracellular regulated kinase), which then dimerizes and translocates to the nucleus where it activates transcription factors such as Elk-1 to modify polynucleotide expression. MEK/ERK kinases have been shown to impair replication of Salmonella within macrophages. Signal transduction by MEK kinase and NADPH oxidase may play an important role in innate host defense against intracellular pathogens. By affecting the MAP kinases as shown below the cationic peptides have an effect on bacterial infection. The cationic peptides can directly affect kinases. Table 21 demonstrates but is not limited to MAP kinase polynucleotide expression changes in response to peptide. The kinases include MAP kinase kinase 6 (H070920), MAP kinase kinase 5 (W69649), MAP kinase 7 (H39192), MAP kinase 12 (AI936909) and MAP kinase-activated protein kinase 3 (W6828 1).


In another method, the methods of the invention may be used in combination, to identify an agent with multiple characteristics, i.e. a peptide with anti-inflammatory/anti-sepsis activity, and the ability to enhance innate immunity, in part by inducing chemokines in vivo.


In another aspect, the invention provides a method for inferring a state of infection in a mammalian subject from a nucleic acid sample of the subject by identifying in the nucleic acid sample a polynucleotide expression pattern exemplified by an increase in polynucleotide expression of at least 2 polynucleotides in Table 55 as compared to a non-infected subject. In another aspect the invention provides a method for inferring a state of infection in a mammalian subject from a nucleic acid sample of the subject by identifying in the nucleic acid sample a polynucleotide expression pattern exemplified by a polynucleotide expression of at least 2 polynucleotides in Table 56 or Table 57 as compared to a non-infected subject. In one aspect of the invention, the state of infection is due to infectious agents or signaling molecules derived therefrom, such as, but not limited to, Gram negative bacteria and Gram positive bacteria, viral, fungal or parasitic agents. In still another aspect the invention provides a polynucleotide expression pattern of a subject having a state of infection identified by the above method. Once identified, such polynucleotides will be useful in methods of diagnosis of a condition associated with the activity or presence of such infectious agents or signaling molecules.


Example 10 below demonstrates this aspect of the invention. Specifically, table 61 demonstrates that both MEK and the NADPH oxidase inhibitors can limit bacterial replication (infection of IFN-γ-primed macrophages by S. typhimurium triggers a MEK kinase). This is an example of how bacterial survival can be impacted by changing host cell signaling molecules.


In still another aspect of the invention, compounds are presented that inhibit stromal derived factor-1 (SDF-1) induced chemotaxis of T cells. Compounds are also presented which decrease expression of SDF-1 receptor. Such compounds also may act as an antagonist or inhibitor of CXCR-4. In one aspect the invention provides a cationic peptide that is an antagonist of CXCR-4. In another aspect the invention provides a method of identifying a cationic peptide that is an antagonist of CXCR-4. The method includes contacting T cells with SDF-1 in the presence of absence of a test peptide and measuring chemotaxis. A decrease in chemotaxis in the presence of the test peptide is then indicative of a peptide that is an antagonist of CXCR-4. Such compounds and methods are useful in therapeutic applications in HIV patients. These types of compounds and the utility thereof is demonstrated, for example, in Example 11 (see also Tables 62, 63). In that example, cationic peptides are shown to inhibit cell migration and therefore antiviral activity.


In one embodiment, the invention provides an isolated cationic peptides having an amino acid sequence of the general formula (Formula A): X1X2X3IX4PX4IPX5X2X1 (SEQ ID NO: 4), wherein X1 is one or two of R, L or K, X2 is one of C, S or A, X3 is one of R or P, X4 is one of A or V and X5 is one of V or W. Examples of-the peptides of the invention include, but are not limited to: LLCRIVPVIPWCK (SEQ ID NO: 5), LRCPIAPVIPVCKK (SEQ ID NO: 6), KSRIVPAIPVSLL (SEQ ID NO: 7), KKSPIAPAIPWSR (SEQ ID NO: 8), RRARIVPAIPVARR (SEQ ID NO: 9) and LSRIAPAIPWAKL (SEQ ID NO: 10).


In another embodiment, the invention provides an isolated linear cationic peptide having an amino acid sequence of the general formula (Formula B):


X1LX2X3KX4X2X5X3PX3X1 (SEQ ID NO: 11), wherein XI is one or two of D, E, S, T or N, X2 is one or two of P, G or D, X3 is one of G, A, V, L, I or Y, X4 is one of R, K or H and X5 is one of S, T, C, M or R. Examples of the peptides of the invention include, but are not limited to:

DLPAKRGSAPGST,(SEQ ID NO: 12)SELPGLKHPCVPGS,(SEQ ID NO: 13)TTLGPVKRDSIPGE,(SEQ ID NO: 14)SLPIKHDRLPATS,(SEQ ID NO: 15)ELPLKRGRVPVE(SEQ ID NO: 16)andNLPDLKKPRVPATS.(SEQ ID NO: 17)


In another embodiment, the invention provides an isolated linear cationic peptide having an amino acid sequence of the general formula (Formula C): X1X2X3X4WX4WX4X5K (SEQ ID NO: 18) (this formula includes CP12a and CP12d), wherein X1 is one to four chosen from A, P or R, X2 is one or two aromatic amino acids (F, Y and W), X3 is one of P or K, X4 is one, two or none chosen from A, P, Y or W and X5 is one to three chosen from R or P. Examples of the peptides of the invention include, but are not limited to:

RPRYPWWPWWPYRPRK,(SEQ ID NO: 19)RRAWWKAWWARRK,(SEQ ID NO: 20)RAPYWPWAWARPRK,(SEQ ID NO: 21)RPAWKYWWPWPWPRRK,(SEQ ID NO: 22)RAAFKWAWAWWRRK(SEQ ID NO: 23)andRRRWKWAWPRRK.(SEQ ID NO: 24)


In another embodiment, the invention provides an isolated hexadecameric cationic peptide having an amino acid sequence of the general formula (Formula D):


X1X2X3X4X1VX3X4RGX4X3X4X1X3X1 (SEQ ID NO: 25) wherein X1 is one or two of R or K, X2 is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3 is C, S, M, D or A and X4 is F, I, V, M or R. Examples of the peptides of the invention include, but are not limited to: RRMCIKVCVRGVCRRKCRK (SEQ ID NO: 26), KRSCFKVSMRGVSRRRCK (SEQ ID NO: 27), KKDAIKKVDIRGMDMRRAR (SEQ ID NO: 28), RKMVKVDVRGIMIRKDRR (SEQ ID NO: 29), KQCVKVAMRGMALRRCK (SEQ ID NO: 30) and RREAIRRVAMRGRDMKRMRR (SEQ ID NO: 31).


In still another embodiment, the invention provides an isolated hexadecameric cationic peptide having an amino acid sequence of the general formula (Formula E): X1X2X3X4X1VX5X4RGX4X5X4X1X3X1 (SEQ ID NO: 32), wherein XI is one or two of R or K, X2 is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3 is one of C, S, M, D or A, X4 is one of F, I, V, M or R and X5 is one of A, I, S, M, D or R. Examples of the peptides of the invention include, but are not limited to: RTCVKRVAMRGIIRKRCR (SEQ ID NO: 33), KKQMMKRVDVRGISVKRKR (SEQ ID NO: 34), KESIKVIIRGMMVRMKK (SEQ ID NO: 35), RRDCRRVMVRGIDIKAK (SEQ ID NO: 36), KRTAIKKVSRRGMSVKARR (SEQ ID NO: 37) and RHCIRRVSMRGIIMRRCK (SEQ ID NO: 38).


In another embodiment, the invention provides an isolated longer cationic peptide having an amino acid sequence of the general formula (Formula F): KX1KX2FX2KMLMX2ALKKX3 (SEQ ID NO: 39), wherein X1 is a polar amino acid (C, S, T, M, N and Q); X2 is one of A, L, S or K and X3 is 1-17 amino acids chosen from G, A, V, L, I, P, F, S, T, K and H. Examples of the peptides of the invention include, but are not limited to:

KCKLFKKMLMLALKKVLTTGLPALKLTK,(SEQ ID NO: 40)KSKSFLKMLMKALKKVLTTGLPALIS,(SEQ ID NO: 41)KTKKFAKMLMMALKKVVSTAKPLAILS,(SEQ ID NO: 42)KMKSFAKMLMLALKKVLKVLTTALTLKAGLPS,(SEQ ID NO: 43)KNKAFAKMLMKALKKVTTAAKPLTG(SEQ ID NO: 44)andKQKLFAKMLMSALKKKTLVTTPLAGK.(SEQ ID NO: 45)


In yet another embodiment, the invention provides an isolated longer cationic peptide having an amino acid sequence of the general formula (Formula G): KWKX2X1X1X2X2X1X2X2XX1X2X2IFHTALKPISS (SEQ ID NO: 46), wherein X1 is a hydrophobic amino acid and X2 is a hydrophilic amino acid. Examples of the peptides of the invention include, but are not limited to:

KWKSFLRTKFSPVRTIFHTALKPISS,(SEQ ID NO: 47)KWKSYAHTIMSPVRLIFHTALKPISS,(SEQ ID NO: 48)KWKRGAHRFMKFLSTIFHTALKPISS,(SEQ ID NO: 49)KWKKWAHSPRKVLTRIFHTALKPISS,(SEQ ID NO: 50)KWKSLVMMFKKPARRIFHTALKPISS(SEQ ID NO: 51)andKWKHALMKAHMLWHMIFHTALKPISS.(SEQ ID NO: 52)


In still another embodiment, the invention provides an isolated cationic peptide having an amino acid sequence of the formula: KWKSFLRTFKSPVRTVFHTALKPISS (SEQ ID NO: 53) or KWKSYAHTIMSPVRLVFHTALKPISS (SEQ ID NO: 54).


The termn “isolated” as used herein refers to a peptide that is substantially free of other proteins, lipids, and nucleic acids (e.g., cellular components with which an in vivo-produced peptide would naturally be associated). Preferably, the peptide is at least 70%, 80%, or most preferably 90% pure by weight and when assessed in exclusion of counter-ion.


The invention also includes analogs, derivatives, conservative variations, and cationic peptide variants of the enumerated polypeptides, provided that the analog, derivative, conservative variation, or variant has a detectable activity in which it enhances innate immunity or has anti-inflammatory activity. It is not necessary that the analog, derivative, variation, or variant have activity identical to the activity of the peptide from which the analog, derivative, conservative variation, or variant is derived.


A cationic peptide “variant” is a peptide that is an altered form of a referenced cationic peptide. For example, the term “variant” includes a cationic peptide in which at least one amino acid of a reference peptide is substituted in an expression library. The term “reference” peptide means any of the cationic peptides of the invention (e.g. as defined in the above formulas), from which a variant, derivative, analog, or conservative variation is derived. Included within the term “derivative” is a hybrid peptide that includes at least a portion of each of two cationic peptides (e.g., 30-80% of each of two cationic peptides). Also included are peptides in which one or more amino acids are deleted from the sequence of a peptide enumerated herein, provided that the derivative has activity in which it enhances innate immunity or has anti-inflammatory activity. This can lead to the development of a smaller active molecule which would also have utility. For example, amino or carboxy terminal amino acids which may not be required for enhancing innate immunity or anti-inflammatory activity of a peptide can be removed. Likewise, additional derivatives can be produced by adding one or a few (e.g., less than 5) amino acids to a cationic peptide without completely inhibiting the activity of the peptide. In addition, C-terminal derivatives, e.g., C-terminal methyl esters, and N-terminal derivatives can be produced and are encompassed by the invention. Peptides of the invention include any analog, homolog, mutant, isomer or derivative of the peptides disclosed in the present invention, so long as the bioactivity as described herein remains. Also included is the reverse sequence of a peptide encompassed by the general formulas set forth above. Additionally, an amino acid of “D” configuration may be substituted with an amino acid of “L” configuration and vice versa. Alternatively the peptide may be cyclized chemically or by the addition of two or more cysteine residues within the sequence and oxidized to form disulphide bonds.


The invention also includes peptides that are conservative variations of those peptides exemplified herein. The term “conservative variation” as used herein denotes a polypeptide in which at least one amino acid is replaced by another, biologically similar residue. Examples of conservative variations include the substitution of one hydrophobic residue, such as isoleucine, valine, leucine, alanine, cysteine, glycine, phenylalanine, proline, tryptophan, tyrosine, norleucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like. Neutral hydrophilic amino acids that can be substituted for one another include asparagine, glutamine, serine and threonine. The term “conservative variation” also encompasses a peptide having a substituted amino acid in place of an unsubstituted parent amino acid. Such substituted amino acids may include amino acids that have been methylated or amidated. Other substitutions will be known to those of skill in the art. In one aspect, antibodies raised to a substituted polypeptide will also specifically bind the unsubstituted polypeptide.


Peptides of the invention can be synthesized by commonly used methods such as those that include t-BOC or FMOC protection of alpha-amino groups. Both methods involve stepwise synthesis in which a single amino acid is added at each step starting from the C-terminus of the peptide (See, Coligan, et al., Current Protocols in Immunology, Wiley Interscience, 1991, Unit 9). Peptides of the invention can also be synthesized by the well known solid phase peptide synthesis methods such as those described by Merrifield, J. Am. Chem. Soc., 85:2149, 1962) and Stewart and Young, Solid Phase Peptides Synthesis, Freeman, San Francisco, 1969, pp. 27-62) using a copoly(styrene-divinylbenzene) containing 0.1-1.0 mMol amines/g polymer. On completion of chemical synthesis, the peptides can be deprotected and cleaved from the polymer by treatment with liquid HF-10% anisole for about ¼-1 hours at 0° C. After evaporation of the reagents, the peptides are extracted from the polymer with a 1% acetic acid solution, which is then lyophilized to yield the crude material. The peptides can be purified by such techniques as gel filtration on Sephadex G-15 using 5% acetic acid as a solvent. Lyophilization of appropriate fractions of the column eluate yield homogeneous peptide, which can then be characterized by standard techniques such as amino acid analysis, thin layer chromatography, high performance liquid chromatography, ultraviolet absorption spectroscopy, molar rotation, or measuring solubility. If desired, the peptides can be quantitated by the solid phase Edman degradation.


The invention also includes isolated nucleic acids (e.g., DNA, cDNA, or RNA) encoding the peptides of the invention. Included are nucleic acids that encode analogs, mutants, conservative variations, and variants of the peptides described herein. The term “isolated” as used herein refers to a nucleic acid that is substantially free of proteins, lipids, and other nucleic acids with which an in vivo-produced nucleic acids naturally associated. Preferably, the nucleic acid is at least 70%, 80%, or preferably 90% pure by weight, and conventional methods for synthesizing nucleic acids in vitro can be used in lieu of in vivo methods. As used herein, “nucleic acid” refers to a polymer of deoxyribo-nucleotides or ribonucleotides, in the form of a separate fragment or as a component of a larger genetic construct (e.g., by operably linking a promoter to a nucleic acid encoding a peptide of the invention). Numerous genetic constructs (e.g., plasmids and other expression vectors) are known in the art and can be used to produce the peptides of the invention in cell-free systems or prokaryotic or eukaryotic (e.g., yeast, insect, or mammalian) cells. By taking into account the degeneracy of the genetic code, one of ordinary skill in the art can readily synthesize nucleic acids encoding the polypeptides of the invention. The nucleic acids of the invention can readily be used in conventional molecular biology methods to produce the peptides of the invention.


DNA encoding the cationic peptides of the invention can be inserted into an “expression vector.” The term “expression vector” refers to a genetic construct such as a plasmid, virus or other vehicle known in the art that can be engineered to contain a nucleic acid encoding a polypeptide of the invention. Such expression vectors are preferably plasmids that contain a promoter sequence that facilitates transcription of the inserted genetic sequence in a host cell. The expression vector typically contains an origin of replication, and a promoter, as well as polynucleotides that allow phenotypic selection of the transformed cells (e.g., an antibiotic resistance polynucleotide). Various promoters, including inducible and constitutive promoters, can be utilized in the invention. Typically, the expression vector contains a replicon site and control sequences that are derived from a species compatible with the host cell.


Transformation or transfection of a recipient with a nucleic acid of the invention can be carried out using conventional techniques well known to those skilled in the art. For example, where the host cell is E. coli, competent cells that are capable of DNA uptake can be prepared using the CaCI2, MgCl2 or RbCl methods known in the art. Alternatively, physical means, such as electroporation or microinjection can be used. Electroporation allows transfer of a nucleic acid into a cell by high voltage electric impulse. Additionally, nucleic acids can be introduced into host cells by protoplast fusion, using methods well known in the art. Suitable methods for transforming eukaryotic cells, such as electroporation and lipofection, also are known.


“Host cells” or “Recipient cells” encompassed by of the invention are any cells in which the nucleic acids of the invention can be used to express the polypeptides of the invention. The term also includes any progeny of a recipient or host cell. Preferred recipient or host cells of the invention include E. coli, S. aureus and P. aeruginosa, although other Gram-negative and Gram-positive bacterial, fungal and mammalian cells and organ isms known in the art can be utilized as long as the expression vectors contain an origin of replication to permit expression in the host.


The cationic peptide polynucleotide sequence used according to the method of the invention can be isolated from an organism or syrithesized in the laboratory. Specific DNA sequences encoding the cationic peptide of interest can be obtained by: 1) isolation of a double-stranded DNA sequence from the genomic DNA; 2) chemical manufacture of a DNA sequence to provide the necessary codons for the cationic peptide of interest; and 3) in vitro synthesis of a double-stranded DNA sequence by reverse transcription of mRNA isolated from a donor cell. In the latter case, a double-stranded DNA complement of mRNA is eventually fo)rmed which is generally referred to as cDNA.


The synthesis of DNA sequences is frequently the method of choice when the entire sequence of amino acid residues of the desired peptide product is known. In the present invention, the synthesis of a DNA sequence has the advantage of allowing the incorporation of codons which are more likely to be recognized by a bacterial host, thereby permitting high level expression without difficulties in translation. In addition, virtually any peptide can be synthesized, including those encoding natural cationic peptides, variants of the same, or synthetic peptides.


When the entire sequence of the desired peptide is not known, the direct synthesis of DNA sequences is not possible and the method of choice is the formation of cDNA sequences. Among the standard procedures for isolating cDNA sequences of interest is the formation of plasmid or phage containing cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expression. When used in combination with polymerase chain reaction technology, even rare expression products can be cloned. In those cases where significant portions of the amino acid sequence of the cationic peptide are known, the production of labeled single or double-stranded DNA or RNA probe sequences duplicating a sequence putatively present in the target cDNA may be employed in DNA/DNA hybridization procedures which are carried out on cloned copies of the cDNA which have been denatured into a single stranded form (Jay, et al., Nuc. Acid Res., 11:2325, 1983).


The peptide of the invention can be administered parenterally by injection or by gradual infusion over time. Preferably the peptide is administered in a therapeutically effective amount to enhance or to stimulate an innate immune response. Innate immunity has been described herein, however examples of indicators of stimulation of innate immunity include but are not limited to monocyte activation, proliferation, differentiation or MAP kinase pathway activation.


The peptide can be administered intravenously, intraperitoneally, intramuscularly, subcutaneously, intracavity, or transdermally. Preferred methods for delivery of the peptide include orally, by encapsulation in microspheres or proteinoids, by aerosol delivery to the lungs, or transdermally by iontophoresis or transdermal electroporation. Other methods of administration will be known to those skilled in the art.


Preparations for parenteral administration of a peptide of the invention include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.


In one embodiment, the invention provides a method for synergistic therapy. For example, peptides as described herein can be used in synergistic combination with sub-inhibitory concentrations of antibiotics. Examples of particular classes of antibiotics useful for synergistic therapy with the peptides of the invention include aminoglycosides (e.g., tobramycin), penicillins (e.g., piperacillin), cephalosporins (e.g., ceftazidime), fluoroquinolones (e.g., ciprofloxacin), carbapenems (e.g., imipenem), tetracyclines and macrolides (e.g., erythromycin and clarithromycin). Further to the antibiotics listed above, typical antibiotics include aminoglycosides (amikacin, gentamicin, kanamycin, netilmicin, tobramycin, s-treptomycin, azithromycin, clarithromycin, erythromycin, erythromycin estolate/ethyl-succinate/gluceptate/lactobionate/stearate), beta-lactams such as penicillins (e.g., penicillin G, penicillin V, methicillin, nafcillin, oxacillin, cloxacillin, dicloxacillin, ampicillin, amoxicillin, ticarcillin, carbenicillin, mezlocillin, azlocillin and piperacillin), or cephalosporins (e.g., cephalothin, cefazolin, cefaclor, cefamandole, cefoxitin, cefiuroxime, cefonicid, cefmnetazole, cefotetan, cefprozil, loracarbef, cefetamet, cefoperazone, cefotaxime, ceftizoxime, ceftriaxone, ceftazidime, cefepime, cefixime, cefpodoxime, and cefsulodin). Other classes of antibiotics include carbapenems (e.g., imipenem), monobactams (e.g.,aztreonam), quinolones (e.g., fleroxacin, nalidixic acid, norfloxacin, ciprofloxacin, ofloxacin, enoxacin, lomefloxacin and cinoxacin), tetracyclines (e.g., doxycycline, minocycline, tetracycline), and glycopeptides (e.g., vancomycin, teicoplanin), for example. Other antibiotics include chloramphenicol, clindamycin, trimethoprim, sulfamethoxazole, nitrofurantoin, rifampin, mupirocin and the cationic peptides.


The efficacy of peptides was evaluated therapeutically alone and in combination with sub-optimal concentrations of antibiotics in models of infection. S. aureus is an important Gram positive pathogen and a leading cause of antibiotic resistant infections. Briefly, peptides were tested for therapeutic efficacy in the S. aureus infection model by injecting them alone and in combination with sub-optimal doses of antibiotics 6 hours after the onset of infection. This would simulate the circumstances of antibiotic resistance developing during an infection, such that the MIC of the resistant bacterium was too high to permit successful therapy (i.e the antibiotic dose applied was sub-optimal). It was demonstrated that the combination of antibiotic and peptide resulted in improved efficacy and suggests the potential for combination therapy (see Example 12).


The invention will now be described in greater detail by reference to the following non-limiting examples. While the invention has been described in detail with reference to certain preferred embodiments thereof, it will be understood that modifications and variations are within the spirit and scope of that which is described and claimed.


EXAMPLE 1
Anti-Sepsis/Anti-Inflammatory Activity

Polynucleotide arrays were utilized to determine the effect of cationic peptides on the transcriptional response of epithelial cells. The A549 human epithelial cell line was maintained in DMEM (Gibco) supplemented with 10% fetal bovine serum (FBS, Medicorp). The A549 cells were plated in 100 mm tissue culture dishes at 2.5×106 cells/dish, cultured overnight and then incubated with 100 ng/ml E.coli O111:B4 LPS (Sigma), without (control) or with 50 μg/ml peptide or medium alone for 4 h. After stimulation, the cells were washed once with diethyl pyrocarbonate-treated phosphate buffered saline (PBS), and detached from the dish using a cell scraper. Total RNA was isolated using RNAqueous (Ambion, Austin, Tex.). The RNA pellet was resuspended in RNase-free water containing Superase-In (RNase inhibitor; Ambion). DNA contamination was removed with DNA-free kit, Ambion). The quality of the RNA was assessed by gel electrophoresis on a 1% agarose gel.


The polynucleotide arrays used were the Human Operon arrays (identification number for the genome is PRHU04-S1), which consist of about 14,000 human oligos spotted in duplicate. Probes were prepared from 10 μg of total RNA and labeled with Cy3 or Cy5 labeled dUTP. The probes were purified and hybridized to printed glass slides overnight at 42° C. and washed. After washing, the image was captured using a Perkin Elmer array scanner. The image processing software (Imapolynucleotide 5.0, Marina Del Rey, Calif.) determines the spot mean intensity, median intensities, and background intensities. A “homemade” program was used to remove background. The program calculates the bottom 10% intensity for each subgrid and subtracts this for each grid. Analysis was performed with Genespring software (Redwood City, Calif.). The intensities for each spot were normalized by taking the median spot intensity value from the population of spot values within a slide and comparing this value to the values of all slides in the experiment. The relative changes seen with cells treated with peptide compared to control cells can be found in Tables 1 and 2. These tables reflect only those polynucleotides that demonstrated significant changes in expression of the 14,000 polynucleotides that were tested for altered expression. The data indicate that the peptides have a widespread ability to reduce the expression of polynucleotides that were induced by LPS.


In Table 1, the peptide, SEQ ID NO: 27 is shown to potently reduce the expression of many of the polynucleotides up-regulated by E. coli O1111:B4 LPS as studied by polynucleotide microarrays. Peptide (50 μg/ml) and LPS (0.1 μg/ml) or LPS alone was incubated with the A549 cells for 4 h and the RNA was isolated. Five μg total RNA was used to make Cy3/Cy5 labeled cDNA probes and hybridized onto Human Operon arrays (PRHU04). The intensity of unstimulated cells is shown in the third column of Table 1. The “Ratio: LPS/control” column refers to the intensity of polynucleotide expression in LPS simulated cells divided by in the intensity of unstimulated cells. The “Ratio: LPS+ID 27/control” column refers to the intensity of polynucleotide expression in cells stimulated with LPS and peptide divided by unstimulated cells.

TABLE 1Reduction, by peptide SEQ ID 27, of A549 human epithelial cell polynucleotideexpression up-regulated by E. coli O111:B4 LPSControl:AccessionPolynucleotideMedia onlyRatio:Ratio: LPS + IDNumberaGene FunctionIntensityLPS/control27/controlAL031983Unknown0.032302.85.1L04510ADP-0.655213.61.4ribosylationfactorD87451ring finger3.896183.72.1protein 10AK000869hypothetical0.138120.12.3proteinU78166Ric-like0.05191.70.2expressed inneuronsAJ001403mucin 5 subtype B0.20353.415.9tracheobronchialAB040057serine/threonine0.9544.315.8protein kinaseMASKZ99756Unknown0.14135.914.0L42243interferon0.16327.65.2receptor 2NM_016216RNA lariat6.15122.310.9debranchingenzymeAK001589hypothetical0.64619.21.3proteinAL137376Unknown1.88117.30.6AB007856FEM-1-like2.62715.70.6death receptorbinding proteinAB007854growth arrest-0.84514.82.2specific 7AK000353cytosolic ovarian0.45313.51.0carcinomaantigen 1D14539myeloid/lymphoid2.03311.63.1or mixed-lineage leukemiatranslocated to 1X76785integration site0.72811.61.9for Epstein-BarrvirusM54915pim-1 oncogene1.40411.40.6NM_006092caspase0.36911.00.5recruitmentdomain 4J03925integrin_alpha M0.2729.94.2NM_001663ADP-0.4399.71.7ribosylationfactor 6M23379RAS p21 protein0.5679.32.8activatorK02581thymidine kinase3.0998.63.51 solubleU94831transmembrane 93.2657.11.5superfamilymember 1X70394zinc finger1.4636.91.7protein 146AL137614hypothetical0.7056.81.0proteinU43083guanine0.8416.61.6cription factor 1X56777zona pellucida1.4145.01.4glycoprotein 3ANM_013400replication1.2414.92.0initiation regionproteinNM_002309leukemia1.2864.81.9inhibitory factorNM_001940dentatorubral-2.0344.71.2pallidoluysianatrophyU91316cytosolic acyl2.0434.71.4coenzyme AthioesterhydrolaseX76104death-associated1.1184.61.8protein kinase 1AF131838Unknown1.8794.61.4AL050348Unknown8.5024.41.7D42085KIAA0095 gene1.3234.41.2productX92896Unknown1.6754.31.5U26648syntaxin 5A1.594.31.4X85750monocyte to1.014.31.1macrophagedifferentiation-associatedD14043CD164 antigen_sialomucin1.6834.21.0J04513fibroblast growth1.2814.00.9factor 2U19796melanoma-1.6184.00.6associatedantigenAK000087hypothetical1.4593.91.0proteinAK001569hypothetical1.5083.91.2proteinAF189009ubiquilin 21.4483.81.3U60205sterol-C4-methyl1.5693.70.8oxidase-likeAK000562hypothetical1.1663.70.6proteinAL096739Unknown3.663.70.5AK000366hypothetical15.1923.51.0proteinNM_006325RAN member1.2423.51.4RAS oncogenefamilyX51688cyclin A21.7723.31.0U34252aldehyde1.2643.31.2dehydrogenase 9NM_013241FH1/FH21.2643.30.6domain-containingproteinAF112219esterase1.8393.31.1D/formylglutathione hydrolaseNM_016237anaphase-2.713.20.9promotingcomplex subunit 5AB014569KIAA0669 gene2.7623.20.2productAF151047hypothetical3.0623.11.0proteinX92972protein2.6153.11.1phosphatase 6catalytic subunitAF035309proteasome 26S5.6283.11.3subunit ATPase 5U52960SRB7 homolog1.3913.10.8J04058electron-transfer-3.2653.11.2flavoproteinalphapolypeptideM57230interleukin 60.7933.11.0signal transducerU78027galactosidase_alpha3.5193.11.1AK000264Unknown2.5333.00.6X80692mitogen-2.4632.91.3activated proteinkinase 6L25931lamin B receptor2.1862.70.7X13334CD14 antigen0.3932.51.1M32315tumor necrosis0.6392.40.4factor receptorsuperfamilymember 1BNM_004862LPS-induced6.0772.31.1TNF-alpha factorAL050337interferon2.0642.11.0gamma receptor 1
aAll Accession Numbers in Table 1 through Table 64 refer to GenBank Accession Numbers.


In Table 2, the cationic peptides at a concentration of 50 μg/ml were shown to potently reduce the expression of many of the polynucleotides up-regulated by 100 ng/ml E. coli O111:B4 LPS as studied by polynucleotide microarrays. Peptide and LPS or LPS alone was incubated with the A549 cells for 4 h and the RNA was isolated. 5 μg total RNA was used to make Cy3/Cy5 labeled cDNA probes and hybridized onto Human Operon arrays (PRHU04). The intensity of unstimulated cells is shown in the third column of Table 2. The “Ratio: LPS/control” column refers to the intensity of polynucleotide expression in LPS-simulated cells divided by in the intensity of unstimulated cells. The other columns refer to the intensity of polynucleotide expression in cells stimulated with LPS and peptide divided by unstimulated cells.

TABLE 2Human A549 Epithelial Cell Polynucleotide Expression up-regulatedby E. coli O111:B4 LPS and reduced by Cationic Peptides.Ctrl:MediaRatio:Ratio:Ratio:AccessiononlyRatio:LPS + IDLPS + IDLPS + IDNumberGeneIntensityLPS/Ctrl27/Ctrl16/Ctrl22/CtrlAL031983Unknown0.03302.85.066.910.31L04510ADP-0.66213.61.42.443.79ribosylationfactorD87451ring finger3.90183.72.13.684.28proteinAK000869hypothetical0.14120.12.342.572.58proteinU78166Ric like0.0591.70.2016.8821.37X03066MHC class II0.0636.54.9012.130.98DO betaAK001904hypothetical0.0332.85.930.370.37proteinAB037722Unknown0.0321.40.300.302.36AK001589hypothetical0.6519.21.260.020.43proteinAL137376Unknown1.8817.30.641.301.35L19185thioredoxin-0.0616.30.182.150.18dependent per-oxide reductase 1J05068transcobalamin 10.0415.91.784.340.83AB007856FEM-1-like2.6315.70.623.380.96death receptorbinding proteinAK000353cytosolic0.4513.51.021.732.33ovariancarcinoma ag 1X16940smooth muscle0.2111.83.240.052.26enteric actin γ2M54915pim-1 oncogene1.4011.40.631.251.83AL122111hypothetical0.3710.90.211.350.03proteinM95678phospholipase0.227.22.380.051.33C beta 2AK001239hypothetical2.206.41.271.892.25proteinAC004849Unknown0.146.30.072.700.07X06614retinoic acid1.925.50.771.431.03receptor_alphaAB007896putative L-type0.945.31.822.152.41neutral aminoacid transporterAB010894BAI1-0.695.01.381.031.80associatedproteinU52522partner of1.982.91.350.481.38RAC1AK001440hypothetical1.022.70.431.200.01proteinNM_001148ankyrin 2_neuronal0.262.50.820.040.66X07173inter-alpha0.332.20.440.030.51inhibitor H2AF095687brain and0.392.10.480.030.98nasopharyngealcarcinomasusceptibilityproteinNM_016382NK cell0.272.10.810.590.04activationinducing ligandNAILAB023198KIAA09810.392.00.430.810.92protein


EXAMPLE 2
Neutralization of the Stimulation of Immune Cells

The ability of compounds to neutralize the stimulation of immune cells by both Gram-negative and Gram-positive bacterial products was tested. Bacterial products stimulate cells of the immune system to produce inflammatory cytokines and when unchecked this can lead to sepsis. Initial experiments utilized the murine macrophage cell line RAW 264.7, which was obtained from the American Type Culture Collection, (Manassas, Va.), the human epithelial cell line, A549, and primary macrophages derived from the bone marrow of BALB/c mice (Charles River Laboratories, Wilmington, Mass.). The cells from mouse bone marrow were cultured in 150-mm plates in Dulbecco's modified Eagle medium (DMEM; Life Technologies, Burlington, ON) supplemented with 20% FBS (Sigma Chemical Co, St. Louis, Mo.) and 20% L cell-conditioned medium as a source of M-CSF. Once macrophages were 60-80% confluent, they were deprived of L cell-conditioned medium for 14-16 h to render the cells quiescent and then were subjected to treatments with 100 ng/ml LPS or 100 ng/ml LPS+20 μg/ml peptide for 24 hours. The release of cytokines into the culture supernatant was determined by ELISA (R&D Systems, Minneapolis, Minn.). The cell lines, RAW 264.7 and A549, were maintained in DMEM supplemented with 10% fetal calf serum. RAW 264.7 cells were seeded in 24 well plates at a density of 106 cells per well in DMEM and A549 cells were seeded in 24 well plates at a density of 105 cells per well in DMEM and both were incubated at 37° C. in 5% CO2 overnight. DMEM was aspirated from cells grown overnight and replaced with fresh medium. In some experiments, blood from volunteer human donors was collected (according to procedures accepted by UBC Clinical Research Ethics Board, certificate C00-0537) by venipuncture into tubes (Becton Dickinson, Franklin Lakes, N.J.) containing 14.3 USP units heparin/ml blood. The blood was mixed with LPS with or without peptide in polypropylene tubes at 37° C. for 6 h. The samples were centrifuged for 5 min at 2000×g, the plasma was collected and then stored at −20° C. until being analyzed for IL-8 by ELISA (R&D Systems). In the experiments with cells, LPS or other bacterial products were incubated with the cells for 6-24 hr at 37° C. in 5% CO2. S. typhimurium LPS and E. coli 0111:B4 LPS were purchased from Sigma. Lipoteichoic acid (LTA) from S. aureus (Sigma) was resuspended in endotoxin free water (Sigma). The Limulus amoebocyte lysate assay (Sigma) was performed on LTA preparations to confirm that lots were not significantly contaminated by endotoxin. Endotoxin contamination was less than 1 ng/ml, a concentration that did not cause significant cytokine production in the RAW 264.7 cells. Non-capped lipoarabinomannan (AraLAM ) was a gift from Dr. John T. Belisle of Colorado State University. The AraLAM from Mycobacterium was filter sterilized and the endotoxin contamination was found to be 3.75 ng per 1.0 mg of LAM as determined by Limulus Amebocyte assay. At the same time as LPS addition (or later where specifically described), cationic peptides were added at a range of concentrations. The supernatants were removed and tested for cytokine production by ELISA (R&D Systems). All assays were performed at least three times with similar results. To confirm the anti-sepsis activity in vivo, sepsis was induced by intraperitoneal injection of 2 or 3 μg of E. coli O111:B4 LPS in phosphate-buffered saline (PBS; pH 7.2) into galactosamine-sensitized 8- to 10-week-old female CD-1 or BALB/c mice. In experiments involving peptides, 200 μg in 100 μl of sterile water was injected at separate intraperitoneal sites within 10 min of LPS injection. In other experiments, CD-1 mice were injected with 400 μg E. coli 011 I :B4 LPS and 10 min later peptide (200 μg) was introduced by intraperitoneal injection. Survival was monitored for 48 hours post injection.


Hyperproduction of TNF-α has been classically linked to development of sepsis. The three types of LPS, LTA or AraLAM used in this example represented products released by both Gram-negative and Gram-positive bacteria. Peptide, SEQ ID NO: 1, was able to significantly reduce TNF-A production stimulated by S. typhimurium, B. cepacia, and E. coli O111:B4 LPS, with the former being affected to a somewhat lesser extent (Table 3). At concentrations as low as 1 μg/ml of peptide (0.25 nM) substantial reduction of TNF-α production was observed in the latter two cases. A different peptide, SEQ ID NO: 3 did not reduce LPS-induced production of TNF-α in RAW macrophage cells, demonstrating that this is not a uniform and predictable property of cationic peptides. Representative peptides from each Formula were also tested for their ability to affect TNF-α production stimulated by E. coli O111:B4 LPS (Table 4). The peptides had a varied ability to reduce TNF-α production although many of them lowered TNF-α by at least 60%.


At certain concentrations peptides SEQ ID NO: 1 and SEQ ID NO: 2, could also reduce the ability of bacterial products to stimulate the production of IL-8 by an epithelial cell line. LPS is a known potent stimulus of IL-8 production by epithelial cells. Peptides, at low concentrations (1-20 μg/ml), neutralized the IL-8 induction responses of epithelial cells to LPS (Tables 5-7). Peptide SEQ ID 2 also inhibited LPS-induced production of IL-8 in whole human blood (Table 4). Conversely, high concentrations of peptide SEQ ID NO: 1 (50 to 100 μg/ml) actually resulted in increased levels of IL-8 (Table 5). This suggests that the peptides have different effects at different concentrations.


The effect of peptides on inflammatory stimuli was also demonstrated in primary murine. cells, in that peptide SEQ ID NO: 1 significantly reduced TNF-α production (>90%) by bone marrow-derived macrophages from BALB/c mice that had been stimulated with 100 ng/ml E. coli 0111 :B4 LPS (Table 8). These experiments were performed in the presence of serum, which contains LPS-binding protein (LBP), a protein that can mediate the rapid binding of LPS to CD14. Delayed addition of SEQ ID NO: 1 to the supernatants of macrophages one hour after stimulation with 100 ng/ml E. coli LPS still resulted in substantial reduction (70%) of TNF-α production (Table 9).


Consistent with the ability of SEQ ID NO: 1 to prevent LPS-induced production of TNF-α in vitro, certain peptides also protected mice against lethal shock induced by high concentrations of LPS. In some experiments, CD-1 mice were sensitized to LPS with a prior injection of galactosamine. Galactosamine-sensitized mice that were injected with 3 μg of E. coli 0111:B4 LPS were all killed within 4-6 hours. When 200 μg of SEQ ID NO: 1 was injected 15 min after the LPS, 50% of the mice survived (Table 10). In other experiments when a higher concentration of LPS was injected into BALB/c mice with no D-galactosamine, peptide protected 100% compared to the control group in which there was no survival (Table 13). Selected other peptides were also found to be protective in these models (Tables 11,12).


Cationic peptides were also able to lower the stimulation of macrophages by Gram-positive bacterial products such as Mycobacterium non-capped lipoarabinomannan (AraLAM) and S. aureus LTA. For example, SEQ ID NO: 1 inhibited induction of TNF-α in RAW 264.7 cells by the Gram-positive bacterial products, LTA (Table 14) and to a lesser extent AraLAM (Table 15). Another peptide, SEQ ID NO: 2, was also found to reduce LTA-induced TNF-α production by RAW 264.7 cells. At a concentration of 1 μg/ml SEQ ID NO: 1 was able to substantially reduce (>75%) the induction of TNF-α production by 1 μg/ml S. aureus LTA. At 20 μg/ml SEQ ID NO: 1, there was >60% inhibition of AraLAM induced TNF-α. Polymyxin B (PMB) was included as a control to demonstrate that contaminating endotoxin was not a significant factor in the inhibition by SEQ ID NO: 1 of AraLAM induced TNF-α. These results demonstrate that cationic peptides can reduce the pro-inflammatory cytokine response of the immune system to bacterial products.

TABLE 3Reduction by SEQ ID 1 of LPS induced TNF-αproduction in RAW 264.7 cells.Amount of SEQInhibition off TNF-α (%)*ID NO: 1 (μg/ml)B. cepacia LPSE. coli LPSS. typhimurium LPS0.18.5 ± 2.9 0.0 ± 0.60.0 ± 0 123.0 ± 11.436.6 ± 7.5 9.8 ± 6.6555.4 ± 8  65.0 ± 3.631.1 ± 7.01063.1 ± 8  75.0 ± 3.437.4 ± 7.52071.7 ± 5.8 81.0 ± 3.5 58.5 ± 10.55086.7 ± 4.3 92.6 ± 2.573.1 ± 9.1
RAW 264.7 mouse macrophage cells were stimulated with 100 ng/ml S. typhimurium LPS, 100 ng/ml B. cepacia LPS and 100 ng/ml E. coli 0111:B4 LPS in the presence of the indicated concentrations of SEQ ID 1 for 6 hr. The concentrations of TNF-α released into the culture supernatants were determined by ELISA. 100% represents the amount of TNF-α resulting from RAW 264.7 cells incubated with LPS alone for 6 hours (S. typhimurium LPS = 34.5 ± 3.2 ng/ml,
# B. cepacia LPS = 11.6 ± 2.9 ng/ml, and E. coli 0111:B4 LPS = 30.8 ± 2.4 ng/ml). Background levels of TNF-α production by the RAW 264.7 cells cultured with no stimuli for 6 hours resulted in TNF-α levels ranging from 0.037-0.192 ng/ml. The data is from duplicate samples and presented as the mean of three experiments + standard error.









TABLE 4










Reduction by Cationic Peptides of E. coli LPS


induced TNF-α production in RAW 264.7 cells.










Peptide (20 μg/ml)
Inhibition of TNF-α (%)







SEQ ID NO: 5
65.6 ± 1.6 



SEQ ID NO: 6
59.8 ± 1.2 



SEQ ID NO: 7
50.6 ± 0.6 



SEQ ID NO: 8
39.3 ± 1.9 



SEQ ID NO: 9
58.7 ± 0.8 



SEQ ID NO: 10
55.5 ± 0.52



SEQ ID NO: 12
52.1 ± 0.38



SEQ ID NO: 13
62.4 ± 0.85



SEQ ID NO: 14
50.8 ± 1.67



SEQ ID NO: 15
69.4 ± 0.84



SEQ ID NO: 16
37.5 ± 0.66



SEQ ID NO: 17
28.3 ± 3.71



SEQ ID NO: 19
69.9 ± 0.09



SEQ ID NO: 20
66.1 ± 0.78



SEQ ID NO: 21
67.8 ± 0.6 



SEQ ID NO: 22
73.3 ± 0.36



SEQ ID NO: 23
83.6 ± 0.32



SEQ ID NO: 24
60.5 ± 0.17



SEQ ID NO: 26
54.9 ± 1.6 



SEQ ID NO: 27
51.1 ± 2.8 



SEQ ID NO: 28
 56 ± 1.1



SEQ ID NO: 29
 58.9 ± 0.005



SEQ ID NO: 31
60.3 ± 0.6 



SEQ ID NO: 33
62.1 ± 0.08



SEQ ID NO: 34
53.3 ± 0.9 



SEQ ID NO: 35
60.7 ± 0.76



SEQ ID NO: 36
  63 ± 0.24



SEQ ID NO: 37
58.9 ± 0.67



SEQ ID NO: 38
54 ± 1 



SEQ ID NO: 40
  75 ± 0.45



SEQ ID NO: 41
  86 ± 0.37



SEQ ID NO: 42
80.5 ± 0.76



SEQ ID NO: 43
88.2 ± 0.65



SEQ ID NO: 44
44.9 ± 1.5 



SEQ ID NO: 45
44.7 ± 0.39



SEQ ID NO: 47
36.9 ± 2.2 



SEQ ID NO: 48
  64 ± 0.67



SEQ ID NO: 49
86.9 ± 0.69



SEQ ID NO: 53
46.5 ± 1.3 



SEQ ID NO: 54
  64 ± 0.73









RAW 264.7 mouse macrophage cells were stimulated with 100 ng/ml E. coli 0111:B4 LPS in the presence of the indicated concentrations of cationic peptides for 6 h. The concentrations of TNF-α released into the culture supernatants were determined by ELISA. Background levels of TNF-α production by the RAW 264.7 cells cultured with no stimuli for 6 hours resulted in TNF-α levels ranging from 0.037-0.192 ng/ml. The data is from duplicate samples and




# presented as the mean of three experiments + standard deviation.













TABLE 5










Reduction by SEQ ID NO: 1 of LPS induced


IL-8 production in A549 cells.










SEQ ID NO: 1 (μg/ml)
Inhibition of IL-8 (%)














0.1
  1 ± 0.3



1
32 ± 10



10
60 ± 9 



20
47 ± 12



50
40 ± 13



100
0









A549 cells were stimulated with increasing concentrations of SEQ ID 1 in the presence of LPS (100 ng/ml E. coli O111:B4) for 24 hours. The concentration of IL-8 in the culture supernatants was determined by ELISA. The background levels of IL-8 from cells alone was 0.172 ± 0.029 ng/ml. The data is presented as the mean of three experiments + standard error.














TABLE 6










Reduction by SEQ ID NO: 2 of E. coli LPS induced


IL-8 production in A549 cells.










Concentration of SEQ ID NO: 2 (μg/ml)
Inhibition of IL-8 (%)














0.1
6.8 ± 9.6



1
12.8 ± 24.5



10
29.0 ± 26.0



50
39.8 ± 1.6 



100
45.0 ± 3.5 









Human A549 epithelial cells were stimulated with increasing concentrations of SEQ ID NO: 2 in the presence of LPS (100 ng/ml E. coli O111:B4) for 24 hours. The concentration of IL-8 in the culture supernatants was determined by ELISA. The data is presented as the mean of three experiments + standard error.














TABLE 7










Reduction by SEQ ID NO: 2 of E. coli LPS


induced IL-8 in human blood.










SEQ ID NO: 2 (μg/ml)
IL-8 (pg/ml)














0
3205



10
1912



50
1458









Whole human blood was stimulated with increasing concentrations of peptide and E. coli O111:B4 LPS for 4 hr. The human blood samples were centrifuged and the serum was removed and tested for IL-8 by ELISA. The data is presented as the average of 2 donors.














TABLE 8










Reduction by SEQ ID NO: 1 of E. coli LPS induced


TNF-α production in murine bone marrow macrophages.










Production of




TNF-α (ng/ml)









SEQ ID NO: 1 (μg/ml)
6 hours
24 hours












LPS alone
1.1
1.7


 1
0.02
0.048


 10
0.036
0.08


100
0.033
0.044


No LPS control
0.038
0.06







BALB/c Mouse bone marrow-derived macrophages were cultured for either 6 h or 24 h with 100 ng/ml E. coli 0111:B4 LPS in the presence or absence of 20 μg/ml of peptide. The supernatant was collected and tested for levels of TNF-α by ELISA. The data represents the amount of TNF-α resulting from duplicate wells of bone marrow-derived macrophages incubated with LPS alone for 6 h (1.1 ± 0.09 ng/ml) or 24 h (1.7 ± 0.2 ng/ml). Background levels of TNF-α were



# 0.038 ± 0.008 ng/ml for 6 h and 0.06 ± 0.012 ng/ml for 24 h.













TABLE 9










Inhibition of E. coli LPS-induced TNF-α production


by delayed addition of SEQ ID NO: 1 to A549 cells.










Time of addition of SEQ ID NO: 1




after LPS (min)
Inhibition of TNF-α (%)














0
98.3 ± 0.3



15
89.3 ± 3.8



30
  83 ± 4.6



60
68 ± 8



90
53 ± 8









Peptide (20 μg/ml) was added at increasing time points to wells already containing A549 human epithelial cells and 100 ng/ml E. coli 0111:B4 LPS. The supernatant was collected after 6 hours and tested for levels of TNF-α by ELISA. The data is presented as the mean of three experiments ± standard error.














TABLE 10










Protection against lethal endotoxemia in galactosamine-


sensitized CD-1 mice by SEQ ID NO: 1.











D-Galactosamine

E. coli

Peptide or
Total
Survival post


treatment
0111:B4 LPS
buffer
mice
endotoxin shock















0

3 μg
PBS
5
 5 (100%)


20
mg
3 μg
PBS
12
0 (0%)


20
mg
3 μg
SEQ ID
12
 6 (50%)





NO: 1







CD-1 mice (9 weeks-old) were sensitized to endotoxin by three intraperitoneal injections of galactosamine (20 mg in 0.1 ml sterile PBS). Then endotoxic shock was induced by intraperitoneal injection of E. coli 0111:B4 LPS (3 μg in 0.1 ml PBS). Peptide, SEQ ID NO: 1, (200 μg/mouse = 8 mg/kg) was injected at a separate intraperitoneal site 15 min after injection of LPS. The mice were monitored for 48 hours and the results were recorded.














TABLE 11










Protection against lethal endotoxemia in galactosamine-sensitized


CD-1 mice by Cationic Peptides.














E. coli 0111:B4

Number
Survival



Peptide Treatment
LPS added
of Mice
(%)







Control (no peptide)
2 μg
5
0



SEQ ID NO: 6
2 μg
5
40



SEQ ID NO: 13
2 μg
5
20



SEQ ID NO: 17
2 μg
5
40



SEQ ID NO: 24
2 μg
5
0



SEQ ID NO: 27
2 μg
5
20









CD-1 mice (9 weeks-old) were sensitized to endotoxin by intraperitoneal injection of galactosamine (20 mg in 0.1 ml sterile PBS). Then endotoxic shock was induced by intraperitoneal injection of E. coli 0111:B4 LPS (2 μg in 0.1 ml PBS). Peptide (200 μg/mouse = 8 mg/kg) was injected at a separate intraperitoneal site 15 min after injection of LPS. The mice were monitored for 48 hours and the results were recorded.














TABLE 12










Protection against lethal endotoxemia in galactosamine-sensitized


BALB/c mice by Cationic Peptides.














E. coli

Number
Survival



Peptide Treatment
0111:B4 LPS added
of Mice
(%)
















No peptide
2 μg
10
10



SEQ ID NO: 1
2 μg
6
17



SEQ ID NO: 3
2 μg
6
0



SEQ ID NO: 5
2 μg
6
17



SEQ ID NO: 6
2 μg
6
17



SEQ ID NO: 12
2 μg
6
17



SEQ ID NO: 13
2 μg
6
33



SEQ ID NO: 15
2 μg
6
0



SEQ ID NO: 16
2 μg
6
0



SEQ ID NO: 17
2 μg
6
17



SEQ ID NO: 23
2 μg
6
0



SEQ ID NO: 24
2 μg
6
17



SEQ ID NO: 26
2 μg
6
0



SEQ ID NO: 27
2 μg
6
50



SEQ ID NO: 29
2 μg
6
0



SEQ ID NO: 37
2 μg
6
0



SEQ ID NO: 38
2 μg
6
0



SEQ ID NO: 41
2 μg
6
0



SEQ ID NO: 44
2 μg
6
0



SEQ ID NO: 45
2 μg
6
0









BALB/c mice (8 weeks-old) were sensitized to endotoxin by intraperitoneal injection of galactosamine (20 mg in 0.1 ml sterile PBS). Then endotoxic shock was induced by intraperitoneal injection of E. coli 0111:B4 LPS (2 μg in 0.1 ml PBS). Peptide (200 μg/mouse = 8 mg/kg) was injected at a separate intraperitoneal site 15 min after injection of LPS. The mice were monitored




# for 48 hours and the results were recorded.













TABLE 13










Protection against lethal endotoxemia in BALB/c mice by SEQ ID


NO: 1.














E. coli

Number




Peptide Treatment
0111:B4 LPS
of Mice
Survival (%)
















No peptide
400 μg
5
0



SEQ ID NO: 1
400 μg
5
100









BALB/c mice were injected intraperitoneal with 400 μg E. coli 0111:B4 LPS. Peptide (200 μg/mouse = 8 mg/kg) was injected at a separate intraperitoneal site and the mice were monitored for 48 hours and the results were recorded.














TABLE 14










Peptide inhibition of TNF-α production induced by S. aureus LTA.










SEQ ID NO: 1 added (μg/ml)
Inhibition of TNF-α (%)














0.1
44.5 ± 12.5



1
76.7 ± 6.4 



5
91 ± 1 



10
94.5 ± 1.5 



20
96 ± 1 









RAW 264.7 mouse macrophage cells were stimulated with 1 μg/ml S. aureus LTA in the absence and presence of increasing concentrations of peptide. The supernatant was collected and tested for levels of TNF-α by ELISA. Background levels of TNF-α production by the RAW 264.7 cells cultured with no stimuli for 6 hours resulted in TNF-α levels ranging from 0.037-0.192 ng/ml. The data is presented as the mean of three or more experiments + standard error.














TABLE 15










Peptide inhibition of TNF-α production induced by Mycobacterium


non-capped lipoarabinomannan.










Peptide (20 μg/ml)
Inhibition of TNF-α (%)







No peptide
0



SEQ ID NO: 1
  64 ± 5.9



Polymyxin B
15 ± 2









RAW 264.7 mouse macrophage cells were stimulated with 1 μg/ml AraLAM in the absence and presence of 20 μg/ml peptide or Polymyxin B. The supernatant was collected and tested for levels of TNF-α by ELISA. Background levels of TNF-α production by the RAW 264.7 cells cultured with no stimuli for 6 hours resulted in TNF-α levels ranging from 0.037-0.192 ng/ml. The data is




# presented as the mean inhibition of three or more experiments + standard error.






EXAMPLE 3
Assessment of Toxicity of the Cationic Peptides

The potential toxicity of the peptides was measured in two ways. First, the Cytotoxicity Detection Kit (Roche) (Lactate dehydrogenase-LDH) Assay was used. It is a colorimetric assay for the quantification of cell death and cell lysis, based on the measurement of LDH activity released from the cytosol of damaged cells into the supernatant. LDH is a stable cytoplasmic enzyme present in all cells and it is released into the cell culture supernatant upon damage of the plasma membrane. An increase in the amount of dead or plasma membrane-damaged cells results in an increase of the LDH enzyme activity in the culture supernatant as measured with an ELISA plate reader, OD490 nm (the amount of color formed in the assay is proportional to the number of lysed cells). In this assay, human bronchial epithelial cells (I6HBEo14, HBE) cells were incubated with 100 μg of peptide for 24 hours, the supernatant removed and tested for LDH. The other assay used to measure toxicity of the cationic peptides was the WST-1 assay (Roche). This assay is a colorimetric assay for the quantification of cell proliferation and cell viability, based on the cleavage of the tetrazolium salt WST-1 by mitochondrial dehydrogenases in viable cells (a non-radioactive alternative to the [3H]-thymidine incorporation assay). In this assay, HBE cells were incubated with 100 μg of peptide for 24 hours, and then 10 μl/well Cell Proliferation Reagent WST-1 was added. The cells are incubated with the reagent and the plate is then measured with an ELISA plate reader, OD490 nm.


The results shown below in Tables 16 and 17 demonstrate that most of the peptides are not toxic to the cells tested. However, four of the peptides from Formula F (SEQ ID NOS: 40, 41, 42 and 43) did induce membrane damage as measured by both assays.

TABLE 16Toxicity of the Cationic Peptides as Measured by the LDH ReleaseAssay.TreatmentLDH Release (OD490 nm)No cells Control0.6 ± 0.1Triton X-100 Control4.6 ± 0.1No peptide control 1.0 ± 0.05SEQ ID NO: 11.18 ± 0.05SEQ ID NO: 31.05 ± 0.04SEQ ID NO: 60.97 ± 0.02SEQ ID NO: 71.01 ± 0.04SEQ ID NO: 9 1.6 ± 0.03SEQ ID NO: 101.04 ± 0.04SEQ ID NO: 130.93 ± 0.06SEQ ID NO: 140.99 ± 0.05SEQ ID NO: 160.91 ± 0.04SEQ ID NO: 170.94 ± 0.04SEQ ID NO: 191.08 ± 0.02SEQ ID NO: 201.05 ± 0.03SEQ ID NO: 211.06 ± 0.04SEQ ID NO: 221.29 ± 0.12SEQ ID NO: 231.26 ± 0.46SEQ ID NO: 241.05 ± 0.01SEQ ID NO: 260.93 ± 0.04SEQ ID NO: 270.91 ± 0.04SEQ ID NO: 280.96 ± 0.06SEQ ID NO: 290.99 ± 0.02SEQ ID NO: 310.98 ± 0.03SEQ ID NO: 331.03 ± 0.05SEQ ID NO: 341.02 ± 0.03SEQ ID NO: 350.88 ± 0.03SEQ ID NO: 360.85 ± 0.04SEQ ID NO: 370.96 ± 0.04SEQ ID NO: 380.95 ± 0.02SEQ ID NO: 402.8 ± 0.5SEQ ID NO: 413.3 ± 0.2SEQ ID NO: 423.4 ± 0.2SEQ ID NO: 434.3 ± 0.2SEQ ID NO: 440.97 ± 0.03SEQ ID NO: 450.98 ± 0.04SEQ ID NO: 471.05 ± 0.05SEQ ID NO: 480.95 ± 0.05SEQ ID NO: 53103 ± 0.06Polymyxin B1.21 ± 0.03
Human HBE bronchial epithelial cells were incubated with 100 μg/ml peptide or Polymyxin B for 24 hours. LDH activity was assayed in the supernatant of the cell cultures. As a control for 100% LDH release, Triton X-100 was added. The data is presented as the mean ± standard deviation. Only peptides SEQ ID 40, 41, 42 and 43 showed any significant toxicity.









TABLE 17










Toxicity of the Cationic Peptides as Measured by the WST-1 Assay.










Treatment
OD490 nm







No cells Control
0.24 ± 0.01



Triton X-100 Control
0.26 ± 0.01



No peptide control
1.63 ± 0.16



SEQ ID NO: 1
1.62 ± 0.34



SEQ ID NO: 3
1.35 ± 0.12



SEQ ID NO: 10
1.22 ± 0.05



SEQ ID NO: 6
1.81 ± 0.05



SEQ ID NO: 7
1.78 ± 0.10



SEQ ID NO: 9
1.69 ± 0.29



SEQ ID NO: 13
1.23 ± 0.11



SEQ ID NO: 14
1.25 ± 0.02



SEQ ID NO: 16
1.39 ± 0.26



SEQ ID NO: 17
1.60 ± 0.46



SEQ ID NO: 19
1.42 ± 0.15



SEQ ID NO: 20
1.61 ± 0.21



SEQ ID NO: 21
1.28 ± 0.07



SEQ ID NO: 22
1.33 ± 0.07



SEQ ID NO: 23
1.14 ± 0.24



SEQ ID NO: 24
1.27 ± 0.16



SEQ ID NO: 26
1.42 ± 0.11



SEQ ID NO: 27
1.63 ± 0.03



SEQ ID NO: 28
1.69 ± 0.03



SEQ ID NO: 29
1.75 ± 0.09



SEQ ID NO: 31
1.84 ± 0.06



SEQ ID NO: 33
1.75 ± 0.21



SEQ ID NO: 34
0.96 ± 0.05



SEQ ID NO: 35
1.00 ± 0.08



SEQ ID NO: 36
1.58 ± 0.05



SEQ ID NO: 37
1.67 ± 0.02



SEQ ID NO: 38
1.83 ± 0.03



SEQ ID NO: 40
0.46 ± 0.06



SEQ ID NO: 41
0.40 ± 0.01



SEQ ID NO: 42
0.39 ± 0.08



SEQ ID NO: 43
0.46 ± 0.10



SEQ ID NO: 44
1.49 ± 0.39



SEQ ID NO: 45
1.54 ± 0.35



SEQ ID NO: 47
1.14 ± 0.23



SEQ ID NO: 48
0.93 ± 0.08



SEQ ID NO: 53
1.51 ± 0.37



Polymyxin B
1.30 ± 0.13









HBE cells were incubated with 100 μg/ml peptide or Polymyxin B for 24 hours and cell viability was tested. The data is presented as the mean ± standard deviation. As a control for 100% LDH release, Triton X-100 was added. Only peptides SEQ ID NOS: 40, 41, 42 and 43 showed any significant toxicity.







EXAMPLE 4
Polynucleotide Regulation by Cationic Peptides

Polynucleotide arrays were utilized to determine the effect of cationic peptides by themselves on the transcriptional response of macrophages and epithelial cells. Mouse macrophage RAW 264.7, Human Bronchial cells (HBE), or A549 human epithelial cells were plated in 150 mm tissue culture dishes at 5.6×106 cells/dish, cultured overnight and then incubated with 50 μg/ml peptide or medium alone for 4 h. After stimulation, the cells were washed once with diethyl pyrocarbonate-treated PBS, and detached from the dish using a cell scraper. Total RNA was isolated using Trizol (Gibco Life Technologies). The RNA pellet was resuspended in RNase-free water containing RNase inhibitor (Ambion, Austin, Tex.). The RNA was treated with DNaseI (Clontech, Palo Alto, Calif.) for 1 h at 37° C. After adding termination mix (0.1 M EDTA [pH 8.0], 1 mg/ml glycogen), the samples were extracted once with phenol:chloroform:isoamyl alcohol (25:24:1), and once with chloroform. The RNA was then precipitated by adding 2.5 volumes of 100% ethanol and 1/10th volume sodium acetate, pH 5.2. The RNA was resuspended in RNase-free water with RNase inhibitor (Ambion) and stored at −70° C. The quality of the RNA was assessed by gel electrophoresis on a 1% agarose gel. Lack of genomic DNA contamination was assessed by using the isolated RNA as a template for PCR amplification with P-actin-specific primers (5′-GTCCCTGTATGCCTCTGGTC-3′ (SEQ ID NO: 55) and 5′-GATGTCACGCACGATTTCC-3′(SEQ ID NO: 56)). Agarose gel electrophoresis and ethidium bromide staining confirmed the absence of an amplicon after 35 cycles.


Atlas cDNA Expression Arrays (Clontech, Palo Alto, Calif.), which consist of 588 selected mouse cDNAs spotted in duplicate on positively charged membranes were used for early polynucleotide array studies (Tables 18 and 19). 32P-radiolabeled cDNA probes prepared from 5 μg total RNA were incubated with the arrays overnight at 71° C. The filters were washed extensively and then exposed to a phosphoimager screen (Molecular Dynamics, Sunnyvale, Calif.) for 3 days at 4° C. The image was captured using a Molecular Dynamics PSI phosphoimager. The hybridization signals were analyzed using Atlaslmage 1.0 Image Analysis software (Clontech) and Excel (Microsoft, Redmond, Wash.). The intensities for each spot were corrected for background levels and normalized for differences in probe labeling using the average values for 5 polynucleotides observed to vary little between the stimulation conditions: β-actin, ubiquitin, ribosomal protein S29, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and Ca2+ binding protein. When the normalized hybridization intensity for a given cDNA was less than 20, it was assigned a value of 20 to calculate the ratios and relative expression.


The next polynucleotide arrays used (Tables 21-26) were the Resgen Human cDNA arrays (identification number for the genome is PRHU03-S3), which consist of 7,458 human cDNAs spotted in duplicate. Probes were prepared from 15-20 μg of total RNA and labeled with Cy3 labeled dUTP. The probes were purified and hybridized to printed glass slides overnight at 42° C. and washed. After washing, the image was captured using a Virtek slide reader. The image processing software (Imagene 4.1, Marina Del Rey, Calif.) determines the spot mean intensity, median intensities, and background intensities. Normalization and analysis was performed with Genespring software (Redwood City, Calif.). Intensity values were calculated by subtracting the mean background intensity from the mean intensity value determined by Imagene. The intensities for each spot were normalized by taking the median spot intensity value from the population of spot values within a slide and comparing this value to the values of all slides in the experiment. The relative changes seen with cells treated with peptide compared to control cells can be found in the Tables below.


The other polynucleotide arrays used (Tables 27-35) were the Human Operon arrays (identification number for the genome is PRHU04-S1), which consist of about 14,000 human oligos spotted in duplicate. Probes were prepared from 10 μg of total RNA and labeled with Cy3 or Cy5 labeled dUTP. In these experiments, A549 epithelial cells were plated in 100 mm tissue culture dishes at 2.5×106 cells/dish. Total RNA was isolated using RNAqueous (Ambion). DNA contamination was removed with DNA-free kit (Ambion). The probes prepared from total RNA were purified and hybridized to printed glass slides overnight at 42° C. and washed. After washing, the image was captured using a Perkin Elmer array scanner. The image processing software (Imagene 5.0, Marina Del Rey, Calif.) determines the spot mean intensity, median intensities, and background intensities. An “in house” program was used to remove background. The program calculates the bottom 10% intensity for each subgrid and subtracts this for each grid. Analysis was performed with Genespring software (Redwood City, Calif.). The intensities for each spot were normalized by taking the median spot intensity value from the population of spot values within a slide and comparing this value to the values of all slides in the experiment. The relative changes seen with cells treated with peptide compared to control cells can be found in the Tables below.


Semi-quantitative RT-PCR was performed to confirm polynucleotide array results. 1 μg RNA samples were incubated with 1 μl oligodT (500 μg/ml) and 1 μl mixed dNTP stock at 1 mM, in a 12 μl volume with DEPC treated water at 65° C. for 5 min in a thermocycler. 4 μl 5× First Strand buffer, 2 μl 0.1M DTT, and 1 μl RNaseOUT recombinant ribonuclease inhibitor (40 units/μl) were added and incubated at 42° C. for 2 min, followed by the addition of 1 μl (200 units) of Superscript II (Invitrogen, Burlington, ON). Negative controls for each RNA source were generated using parallel reactions in the absence of Superscript II. cDNAs were amplified in the presence of 5′ and 3′ primers (1.0 μM), 0.2 mM dNTP mixture, 1.5 mM MgCl, 1 U of Taq DNA polymerase (New England Biolabs, Missisauga, ON), and 1× PCR buffer. Each PCR was performed with a thermal cycler by using 30-40 cycles consisting of 30s of denaturation at 94° C., 30s of annealing at either 52° C. or 55° C. and 40s of extension at 72° C. The number of cycles of PCR was optimized to lie in the linear phase of the reaction for each primer and set of RNA samples. A housekeeping polynucleotide 0-actin was amplified in each experiment to evaluate extraction procedure and to estimate the amount of RNA. The reaction product was visualized by electrophoresis and analyzed by densitometry, with relative starting RNA concentrations calculated with reference to β-actin amplification.


Table 18 demonstrates that SEQ ID NO: 1 treatment of RAW 264.7 cells up-regulated the expression of more than 30 different polynucleotides on small Atlas microarrays with selected known polynucleotides. The polynucleotides up-regulated by peptide, SEQ ID NO: 1, were mainly from two categories: one that includes receptors (growth, chemokine, interleukin, interferon, hormone, neurotransmitter), cell surface antigens and cell adhesion and another one that includes cell-cell communication (growth factors, cytokines, chemokines, interleukin, interferons, hormones), cytoskeleton, motility, and protein turnover. The specific polynucleotides up-regulated included those encoding chemokine MCP-3, the anti-inflammatory cytokine IL-10, macrophage colony stimulating factor, and receptors such as IL-1R-2 (a putative antagonist of productive IL-1 binding to IL-1R1), PDGF receptor B, NOTCH4, LIF receptor, LFA-1, TGFβ receptor 1, G-CSF receptor, and IFNγ receptor. The peptide also up-regulated polynucleotides encoding several metalloproteinases, and inhibitors thereof, including the bone morphogenetic proteins BMP-1, BMP-2, BMP-8a, TIMP2 and TIMP3. As well, the peptide up-regulated specific transcription factors, including JunD, and-the YY and LIM-1 transcription factors, and kinases such as Etk1 and Csk demonstrating its widespread effects. It was also discovered from the polynucleotide array studies that SEQ ID NO: 1 down-regulated at least 20 polynucleotides in RAW 264.7 macrophage cells (Table 19). The polynucleotides down-regulated by peptide included DNA repair proteins and several inflammatory mediators such as MIP-1α, oncostatin M and IL-12. A number of the effects of peptide on polynucleotide expression were confirmed by RT-PCR (Table 20). The peptides, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 19, and SEQ ID NO: 1, and representative peptides from each of the formulas also altered the transcriptional responses in a human epithelial cell line using mid-sized microarrays (7835 polynucleotides). The effect of SEQ ID NO: 1 on polynucleotide expression was compared in 2 human epithelial cell lines, A549 and HBE. Polynucleotides related to the host immune response that were up-regulated by 2 peptides or more by a ratio of 2-fold more than unstimulated cells are described in Table 21. Polynucleotides that were down-regulated by 2 peptides or more by a ratio of 2-fold more than unstimulated cells are described in Table 22. In Table 23 and Table 24, the human epithelial pro-inflammatory polynucleotides that are up- and down-regulated respectively are shown. In Table 25 and Table 26 the anti-inflammatory polynucleotides affected by cationic peptides are shown. The trend becomes clear that the cationic peptides up-regulate the anti-inflammatory response and down-regulate the pro-inflammatory response. It was very difficult to find a polynucleotide related to the anti-inflammatory response that was down-regulated (Table 26). The pro-inflammatory polynucleotides upregulated by cationic peptides were mainly polynucleotides related to migration and adhesion. Of the down-regulated pro-inflammatory polynucleotides, it should be noted that all the cationic peptides affected several toll-like receptor (TLR) polynucleotides, which are very important in signaling the host response to infectious agents. An important anti-inflammatory polynucleotide that was up-regulated by all the peptides is the IL-10 receptor. IL-10 is an important cytokine involved in regulating the pro-inflammatory cytokines. These polynucleotide expression effects were also observed using primary human macrophages as observed for peptide SEQ ID NO: 6 in Tables 27 and 28. The effect of representative. peptides from each of the formulas on human epithelial cell expression of selected polynucleotides (out of 14,000 examined) is shown in Tables 31-37 below. At least 6 peptides from each formula were tested for their ability to alter human epithelial polynucleotide expression and indeed they had a wide range of stimulatory effects. In each of the formulas there were at least 50 polynucleotides commonly up-regulated by each of the peptides in the group.

TABLE 18Polynucleotides up-regulated by peptide, SEQ ID NO: 1, treatment of RAWmacrophage cellsa.Polynucleotide/UnstimulatedRatioAccessionProteinPolynucleotide FunctionIntensitypeptide:UnstimulatedbNumberEtk1Tyrosine-protein kinase2043M68513receptorPDGFRBGrowth factor receptor2425X04367Corticotropin releasing2023X72305factor receptorNOTCH4proto-oncopolynucleotide4818M80456IL-1R2Interleukin receptor2016X59769MCP-3Chemokine5614S71251BMP-1Bone2014L24755morphopolynucleotideticproteinEndothelinReceptor2014U32329b receptorc-retOncopolynucleotide2013X67812precursorLIFRCytokine receptor2012D26177BMP-8aBone2012M97017morphopolynucleotideticproteinZfp92Zinc finger protein 928711U47104MCSFMacrophage colony8511X05010stimulating factor 1GCSFRGranulocyte colony-2011M58288stimulating factor receptorIL-8RBChemokine receptor11210D17630IL-9RInterleukin receptor1126M84746CasCrk-associated substrate316U48853p58/GTAKinase2545M58633CASP2Caspase precursor1295D28492IL-1βInterleukin precursor915M15131precursorSPI2-2Serine protease inhibitor625M64086C5ARChemokine receptor3004S46665L-mycOncopolynucleotide2084X13945IL-10Interleukin1684M37897p19ink4cdk4 and cdk6 inhibitor1474U19597ATOH2Atonal homolog 21134U29086DNAse1DNase874U00478CXCR-4Chemokine receptor364D87747Cyclin D3Cyclin3273U43844IL-7RαInterleukin receptor3173M29697POLADNA polymeraseα2413D17384Tie-2Oncopolynucleotide1933S67051DNL1DNA ligase I1403U04674BADApoptosis protein1223L37296GADD45DNA-damage-inducible883L28177proteinSikSrc-related kinase823U16805integrinα4Integrin23242X53176TGFβR1Growth factor receptor10382D25540LAMR1Receptor10012J02870CrkCrk adaptor protein8532S72408ZFXChromosomal protein6792M32309Cyclin E1Cylcin6712X75888POLD1DNA polymerase subunit6492Z21848Vavproto-oncopolynucleotide6132X64361YY (NF-E1)Transcription factor5932L13968JunDTranscription factor5342J050205Cskc-src kinase4892U05247Cdk7Cyclin-dependent kinase4752U11822MLC1AMyosin light subunit4532M19436isoformERBB-3Receptor4352L47240UBFTranscription factor4052X60831TRAILApoptosis ligand3642U37522LFA-1Cell adhesion receptor3402X14951SLAPSrc-like adaptor protein3152U29056IFNGRInterferon gamma receptor3082M28233LIM-1Transcription factor2952Z27410ATF2Transcription factor2872S76657FSTFollistatin precursor2752Z29532TIMP3Protease inhibitor2592L19622RU49Transcription factor2532U41671IGF-1RαInsulin-like growth factor2182U00182receptorCyclin G2Cyclin2142U95826fynTyrosine-protein kinase1912U70324BMP-2Bone1862L25602morphopolynucleotideticproteinBrn-3.2Transcription factor1742S68377POUKIF1AKinesin family protein1692D29951MRC1Mannose receptor1672Z11974PAI2Protease inhibitor1542X19622BKLFCACCC Box-binding1382U36340proteinTIMP2Protease inhibitor1362X62622MasProto-oncopolynucleotide1312X67735NURR-1Transcription factor1292S53744
The cationic peptides at a concentration of 50 μg/ml were shown to potently induce the expression of several polynucleotides. Peptide was incubated with the RAW cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Atlas arrays. The intensity of unstimulated cells is shown in the third column. The “Ratio Peptide:Unstimulated” column refers to
# the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.
The changes in the normalized intensities of the housekeeping polynucleotides ranged from 0.8-1.2 fold, validating the use of these polynucleotides for normalization. When the normalized hybridization intensity for a given cDNA was less than 20, it was assigned a value of 20 to calculate the ratios and relative expression. The array experiments were repeated 3 times with different RNA
# preparations and the average fold change is shown above. Polynucleotides with a two fold or greater change in relative expression levels are presented.









TABLE 19










Polynucleotides down-regulated by SEQ ID NO: 1 treatment of RAW macrophage


cellsa.











Polynucleotide/

Unstimulated
Ratio
Accession


Protein
Polynucleotide Function
Intensity
peptide:Unstimulated
Number














sodium channel
Voltage-gated ion channel
257
0.08
L36179


XRCC1
DNA repair protein
227
0.09
U02887


ets-2
Oncopolynucleotide
189
0.11
J04103


XPAC
DNA repair protein
485
0.12
X74351


EPOR
Receptor precursor
160
0.13
J04843


PEA 3
Ets-related protein
158
0.13
X63190


orphan receptor
Nuclear receptor
224
0.2
U11688


N-cadherin
Cell adhesion receptor
238
0.23
M31131


OCT3
Transcription factor
583
0.24
M34381


PLCβ
phospholipase
194
0.26
U43144


KRT18
Intermediate filament
318
0.28
M11686



proteins


THAM
Enzyme
342
0.32
X58384


CD40L
CD40 ligand
66
0.32
X65453


CD86
T-lymphocyte antigen
195
0.36
L25606


oncostatin M
Cytokine
1127
0.39
D31942


PMS2 DNA
DNA repair protein
200
0.4
U28724


IGFBP6
Growth factor
1291
0.41
X81584


MIP-1β
Cytokine
327
0.42
M23503


ATBF1
AT motif-binding factor
83
0.43
D26046


nucleobindin
Golgi resident protein
367
0.43
M96823


bcl-x
Apoptosis protein
142
0.43
L35049


uromodulin
glycoprotein
363
0.47
L33406


IL-12 p40
Interleukin
601
0.48
M86671


MmRad52
DNA repair protein
371
0.54
Z32767


Tob1
Antiproliferative factor
956
0.5
D78382


Ung1
DNA repair protein
535
0.51
X99018


KRT19
Intermediate filament
622
0.52
M28698



proteins


PLCγ
phospholipase
251
0.52
X95346


Integrin α6
Cell adhesion receptor
287
0.54
X69902


GLUT1
Glucose transporter
524
0.56
M23384


CTLA4
immunoglobin
468
0.57
X05719



superfamily


FRA2
Fos-related antigen
446
0.57
X83971


MTRP
Lysosome-associated
498
0.58
U34259



protein







The cationic peptides at a concentration of 50 μg/ml were shown to reduce the expression of several polynucleotides. Peptide was incubated with the RAW cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Atlas arrays. The intensity of unstimulated cells is shown in the third column. The “Ratio Peptide:Unstimulated” column refers to the



# intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells. The array experiments were repeated 3 times with different cells and the average fold change is shown below. Polynucleotides with an approximately two fold or greater change in relative expression levels are presented.













TABLE 20










Polynucleotide Expression changes in response to peptide, SEQ ID


NO: 1, could be confirmed by RT-PCR.











Polynucleotide
Array Ratio -*
RT-PCR Ratio -*







CXCR-4
4.0 ± 1.7
4.1 ± 0.9



IL-8RB
9.5 ± 7.6
7.1 ± 1.4



MCP-3
13.5 ± 4.4 
 4.8 ± 0.88



IL-10
4.2 ± 2.1
16.6 ± 6.1 



CD14
0.9 ± 0.1
0.8 ± 0.3



MIP-1B
0.42 ± 0.09
0.11 ± 0.04



XRCC1
0.12 ± 0.01
 0.25 ± 0.093



MCP-1
Not on array
3.5 ± 1.4









RAW 264.7 macrophage cells were incubated with 50 μg/ml of peptide or media only for 4 hours and total RNA isolated and subjected to semi-quantitative RT-PCR. Specific primer pairs for each polynucleotide were used for amplification of RNA. Amplification of β-actin was used as a positive control and for standardization. Densitometric analysis of RT-PCR products was used. The results refer




# to the relative fold change in polynucleotide expression of peptide treated cells compared to cells incubated with media alone. The data is presented as the mean ± standard error of three experiments.













TABLE 21










Polynucleotides up-regulated by peptide treatment of A549 epithelial cellsa.











Unstimulated
Ratio Peptide:Unstimulated
Accession













Polynucleotide/Protein
Intensity
ID 2
ID 3
ID 19
ID 1
Number
















IL-1 R antagonist homolog 1
0.00
3086
1856
870

AI167887


IL-10 R beta
0.53
2.5
1.6
1.9
3.1
AA486393


IL-11 R alpha
0.55
2.4
1.0
4.9
1.8
AA454657


IL-17 R
0.54
2.1
2.0
1.5
1.9
AW029299


TNF R superfamily, member
0.28
18
3.0
15
3.6
AA150416


1B


TNF R superfamily, member 5
33.71
3.0
0.02


H98636


(CD40LR)


TNF R superfamily, member
1.00
5.3
4.50
0.8

AA194983


11b


IL-8
0.55
3.6
17
1.8
1.1
AA102526


interleukin enhancer binding
0.75
1.3
2.3
0.8
4.6
AA894687


factor 2


interleukin enhancer binding
0.41
2.7

5.3
2.5
R56553


factor 1


cytokine inducible SH2-
0.03
33
44
39
46
AA427521


containing protein


IK cytokine, down-regulator of
0.50
3.1
2.0
1.7
3.3
R39227


HLA II


cytokine inducible SH2-
0.03
33
44
39
46
AA427521


containing protein


IK cytokine, down-regulator of
0.50
3.1
2.0
1.7
3.3
R39227


HLA II


small inducible cytokine
1.00
3.9


2.4
AI922341


subfamily A (Cys—Cys),


member 21


TGFB inducible early growth
0.90
2.4
2.1
0.9
1.1
AI473938


response 2


NK cell R
1.02
2.5
0.7
0.3
1.0
AA463248


CCR6
0.14
4.5
7.8
6.9
7.8
N57964


cell adhesion molecule
0.25
4.0
3.9
3.9
5.1
R40400


melanoma adhesion molecule
0.05
7.9
20
43
29.1
AA497002


CD31
0.59
2.7
3.1
1.0
1.7
R22412


integrin, alpha 2 (CD49B,
1.00
0.9
2.4
3.6
0.9
AA463257


alpha 2 subunit of VLA-2


receptor


integrin, alpha 3 (antigen
0.94
0.8
2.5
1.9
1.1
AA424695


CD49C, alpha 3 subunit of


VLA-3 receptor)


integrin, alpha E
0.01
180
120
28
81
AA425451


integrin, beta 1
0.47
2.1
2.1
7.0
2.6
W67174


integrin, beta 3
0.55
2.7
2.8
1.8
1.0
AA037229


integrin, beta 3
0.57
2.6
1.4
1.8
2.0
AA666269


integrin, beta 4
0.65
0.8
2.2
4.9
1.5
AA485668


integrin beta 4 binding protein
0.20
1.7
5.0
6.6
5.3
AI017019


calcium and integrin binding
0.21
2.8
4.7
9.7
6.7
AA487575


protein


disintegrin and
0.46
3.1

2.2
3.8
AA279188


metalloproteinase domain 8


disintegrin and
0.94
1.1
2.3
3.6
0.5
H59231


metalloproteinase domain 9


disintegrin and
0.49
1.5
2.1
3.3
2.2
AA043347


metalloproteinase domain 10


disintegrin and
0.44
1.9
2.3
2.5
4.6
H11006


metalloproteinase domain 23


cadherin 1, type 1, E-cadherin
0.42
8.1
2.2
2.4
7.3
H97778


epithelial)


cadherin 12, type 2 (N-
0.11
13
26
9.5

AI740827


cadherin 2)


protocadherin 12
0.09
14.8
11.5
2.6
12.4
AI652584


protocadherin gamma
0.34
3.0
2.5
4.5
9.9
R89615


subfamily C, 3


catenin (cadherin-associated
0.86
1.2
2.2
2.4

AA025276


protein), delta 1


laminin R 1 (67 kD, ribosomal
0.50
0.4
2.0
4.4
3.0
AA629897


protein SA)


killer cell lectin-like receptor
0.11
9.7
9.0
4.1
13.4
AA190627


subfamily C, member 2


killer cell lectin-like receptor
1.00
3.2
1.0
0.9
1.3
W93370


subfamily C, member 3


killer cell lectin-like receptor
0.95
2.3
1.7
0.7
1.1
AI433079


subfamily G, member 1


C-type lectin-like receptor-2
0.45
2.1
8.0
2.2
5.3
H70491


CSF 3 R
0.40
1.9
2.5
3.5
4.0
AA458507


macrophage stimulating 1 R
1.00
1.7
2.3
0.4
0.7
AA173454


BMP R type IA
0.72
1.9
2.8
0.3
1.4
W15390


formyl peptide receptor 1
1.00
3.1
1.4
0.4

AA425767


CD2
1.00
2.6
0.9
1.2
0.9
AA927710


CD36
0.18
8.2
5.5
6.2
2.5
N39161


vitamin D R
0.78
2.5
1.3
1.1
1.4
AA485226


Human proteinase activated R-2
0.54
6.1
1.9
2.2

AA454652


prostaglandin E receptor 3
0.25
4.1
4.9
3.8
4.9
AA406362


(subtype EP3)


PDGF R beta polypeptide
1.03
2.5
1.0
0.5
0.8
R56211


VIP R 2
1.00
3.1


2.0
AI057229


growth factor receptor-bound
0.51
2.2
2.0
2.4
0.3
AA449831


protein 2


Mouse Mammary Turmor
1.00
6.9

16

W93891


Virus Receptor homolog


adenosine A2a R
0.41
3.1
1.8
4.0
2.5
N57553


adenosine A3 R
0.83
2.0
2.3
1.0
1.2
AA863086


T cell R delta locus
0.77
2.7
1.3

1.8
AA670107


prostaglandin E receptor 1
0.65
7.2

6.0
1.5
AA972293


(subtype EP1)


growth factor receptor-bound
0.34

3.0
6.3
2.9
R24266


protein 14


Epstein-Barr virus induced
0.61
1.6
2.4

8.3
AA037376


polynucleotide 2


complement component
0.22
26
4.5
2.6
18.1
AA521362


receptor 2


endothelin receptor type A
0.07
12
14
14
16
AA450009


v-SNARE R
0.56
11
12
1.8

AA704511


tyrosine kinase, non-receptor, 1
0.12
7.8
8.5
10
8.7
AI936324


receptor tyrosine kinase-like
0.40
7.3
5.0
1.6
2.5
N94921


orphan receptor 2


protein tyrosine phosphatase,
1.02
1.0
13.2
0.5
0.8
AA682684


non-receptor type 3


protein tyrosine phosphatase,
0.28
3.5
4.0
0.9
5.3
AA434420


non-receptor type 9


protein tyrosine phosphatase,
0.42
2.9
2.4
2.2
3.0
AA995560


non-receptor type 11


protein tyrosine phosphatase,
1.00
2.3
2.2
0.8
0.5
AA446259


non-receptor type 12


protein tyrosine phosphatase,
0.58
1.7
2.4
3.6
1.7
AA679180


non-receptor type 13


protein tyrosine phosphatase,
0.52
3.2
0.9
1.9
6.5
AI668897


non-receptor type 18


protein tyrosine phosphatase,
0.25
4.0
2.4
16.8
12.8
H82419


receptor type, A


protein tyrosine phosphatase,
0.60
3.6
3.2
1.6
1.0
AA045326


receptor type, J


protein tyrosine phosphatase,
0.73
1.2
2.8
3.0
1.4
R52794


receptor type, T


protein tyrosine phosphatase,
0.20
6.1
1.2
5.6
5.0
AA644448


receptor type, U


protein tyrosine phosphatase,
1.00
5.1


2.4
AA481547


receptor type, C-associated


protein


phospholipase A2 receptor 1
0.45
2.8
2.2
1.9
2.2
AA086038


MAP kinase-activated protein
0.52
2.1
2.7
1.1
1.9
W68281


kinase 3


MAP kinase kinase 6
0.10
18
9.6

32
H07920


MAP kinase kinase 5
1.00
3.0
5.2
0.8
0.2
W69649


MAP kinase 7
0.09

11.5
12
33
H39192


MAP kinase 12
0.49
2.1
1.7
2.2
2.0
AI936909


G protein-coupled receptor 4
0.40
3.7
3.0
2.4
2.5
AI719098


G protein-coupled receptor 49
0.05

19
19
27
AA460530


G protein-coupled receptor 55
0.08
19
15
12

N58443


G protein-coupled receptor 75
0.26
5.2
3.1
7.1
3.9
H84878


G protein-coupled receptor 85
0.20
6.8
5.4
4.9
5.0
N62306


regulator of G-protein
0.02
48
137
82

AI264190


signaling 20


regulator of G-protein
0.27

3.7
8.9
10.6
R39932


signaling 6


BCL2-interacting killer
1.00
1.9

5.2

AA291323


(apoptosis-inducing)


apoptosis inhibitor 5
0.56
2.8
1.6
2.4
1.8
AI972925


caspase 6, apoptosis-related
0.79
0.7
2.6
1.3
2.8
W45688


cysteine protease


apoptosis-related protein
0.46
2.2
1.4
2.3
2.9
AA521316


PNAS-1


caspase 8, apoptosis-related
0.95
2.2
1.0
0.6
2.0
AA448468


cysteine protease







The cationic peptides at concentrations of 50 μg/ml were shown to increase the expression of several polynucleotides. Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human cDNA arrays ID#PRHU03-S3. The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Unstimulated” columns refers to the intensity



# of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 22










Polynucleotides down-regulated by peptide treatment of A549 epithelial cellsa.











Unstimulated
Ratio Peptide:Unstimulated
Accession













Polynucleotide/Protein
Intensity
ID 2
ID 3
ID 19
ID 1
Number
















TLR 1
3.22
0.35
0.31
0.14
0.19
AI339155


TLR 2
2.09
0.52
0.31
0.48
0.24
T57791


TLR 5
8.01
0.12
0.39


N41021


TLR 7
5.03
0.13
0.11
0.20
0.40
N30597


TNF receptor-associated factor 2
0.82
1.22
0.45
2.50
2.64
T55353


TNF receptor-associated factor 3
3.15
0.15

0.72
0.32
AA504259


TNF receptor superfamily, member 12
4.17
0.59
0.24

0.02
W71984


TNF R superfamily, member 17
2.62

0.38
0.55
0.34
AA987627


TRAF and TNF receptor-associated
1.33
0.75
0.22
0.67
0.80
AA488650


protein


IL-1 receptor, type I
1.39
0.34
0.72
1.19
0.34
AA464526


IL-2 receptor, alpha
2.46
0.41
0.33
0.58

AA903183


IL-2 receptor, gamma (severe
3.34
0.30
0.24

0.48
N54821


combined immunodeficiency)


IL-12 receptor, beta 2
4.58
0.67
0.22


AA977194


IL-18 receptor 1
1.78
0.50
0.42
0.92
0.56
AA482489


TGF beta receptor III
2.42
0.91
0.24
0.41
0.41
H62473


leukotriene b4 receptor (chemokine
1.00

1.38
4.13
0.88
AI982606


receptor-like 1)


small inducible cytokine subfamily A
2.26
0.32

0.44
1.26
AA495985


(Cys—Cys), member 18


small inducible cytokine subfamily A
2.22
0.19
0.38
0.45
0.90
AI285199


(Cys—Cys), member 20


small inducible cytokine subfamily A
2.64
0.38
0.31
1.53

AA916836


(Cys—Cys), member 23


small inducible cytokine subfamily B
3.57
0.11
0.06
0.28
0.38
AI889554


(Cys-X-Cys), member 6 (granulocyte


chemotactic protein 2)


small inducible cytokine subfamily B
2.02
0.50
1.07
0.29
0.40
AA878880


(Cys-X-Cys), member 10


small inducible cytokine A3
2.84
1.79
0.32
0.35

AA677522


(homologous to mouse Mip-1a)


cytokine-inducible kinase
2.70
0.41
0.37
0.37
0.34
AA489234


complement component C1q receptor
1.94
0.46
0.58
0.51
0.13
AI761788


cadherin 11, type 2, OB-cadherin
2.00
0.23
0.57
0.30
0.50
AA136983


(osteoblast)


cadherin 3, type 1, P-cadherin
2.11
0.43
0.53
0.10
0.47
AA425217


(placental)


cadherin, EGF LAG seven-pass. G-type
1.67
0.42
0.41
1.21
0.60
H39187


receptor 2, flamingo (Drosophila)


homolog


cadherin 13, H-cadherin (heart)
1.78
0.37
0.40
0.56
0.68
R41787


selectin L (lymphocyte adhesion
4.43
0.03
0.23
0.61

H00662


molecule 1)


vascular cell adhesion molecule 1
1.40
0.20
0.72
0.77
0.40
H16591


intercellular adhesion molecule 3
1.00
0.12
0.31
2.04
1.57
AA479188


integrin, alpha 1
2.42
0.41
0.26

0.56
AA450324


integrin, alpha 7
2.53
0.57
0.39
0.22
0.31
AA055979


integrin, alpha 9
1.16
0.86
0.05
0.01
2.55
AA865557


integrin, alpha 10
1.00
0.33
0.18
1.33
2.25
AA460959


integrin, beta 5
1.00
0.32
1.52
1.90
0.06
AA434397


integrin, beta 8
3.27
0.10
1.14
0.31
0.24
W56754


disintegrin and metalloproteinase
2.50
0.40
0.29
0.57
0.17
AI205675


domain 18


disintegrin-like and metalloprotease
2.11
0.32
0.63
0.47
0.35
AA398492


with thrombosondin type 1 motif, 3


disintegrin-like and metalloprotease
1.62
0.39
0.42
1.02
0.62
AI375048


with thrombospondin type 1 motif, 5


T-cell receptor interacting molecule
1.00
0.41
1.24
1.41
0.45
AI453185


diphtheria toxin receptor (heparin-
1.62
0.49
0.85
0.62
0.15
R45640


binding epidermal growth factor-like


growth factor


vasoactive intestinal peptide receptor 1
2.31
0.43
0.31
0.23
0.54
H73241


Fc fragment of IgG, low affinity IIIb,
3.85
−0.20
0.26
0.76
0.02
H20822


receptor for (CD16)


Fc fragment of IgG, low affinity IIb,
1.63
0.27
0.06
1.21
0.62
R68106


receptor for (CD32)


Fc fragment of IgE, high affinity I,
1.78
0.43
0.00
0.56
0.84
AI676097


receptor for; alpha polypeptide


leukocyte immunoglobulin-like
2.25
0.44
0.05
0.38
0.99
N63398


receptor, subfamily A


leukocyte immunoglobulin-like
14.21


1.10
0.07
AI815229


receptor, subfamily B (with TM and


ITIM domains), member 3


leukocyte immunoglobulin-like
2.31
0.75
0.43
0.19
0.40
AA076350


receptor, subfamily B (with TM and


ITIM domains), member 4


leukocyte immunoglobulin-like
1.67
0.35
0.60
0.18
0.90
H54023


receptor, subfamily B


peroxisome proliferative activated
1.18
0.38
0.85
0.87
0.26
AI739498


receptor, alpha


protein tyrosine phosphatase, receptor
2.19
0.43

1.06
0.46
N49751


type, f polypeptide (PTPRF),


interacting protein (liprin), α1


protein tyrosine phosphatase, receptor
1.55
0.44
0.64
0.30
0.81
H74265


type, C


protein tyrosine phosphatase, receptor
2.08
0.23
0.37
0.56
0.48
AA464542


type, E


protein tyrosine phosphatase, receptor
2.27
0.02
0.44

0.64
AA464590


type, N polypeptide 2


protein tyrosine phosphatase, receptor
2.34
0.11
0.43
0.24
0.89
AI924306


type, H


protein tyrosine phosphatase, receptor-
1.59
0.63
0.34
0.72
0.35
AA476461


type, Z polypeptide 1


protein tyrosine phosphatase, non-
1.07
0.94
0.43
0.25
1.13
H03504


receptor type 21


MAP kinase 8 interacting protein 2
1.70
0.07
0.85
0.47
0.59
AA418293


MAP kinase kinase kinase 4
1.27
0.37
0.79
1.59
−5.28
AA402447


MAP kinase kinase kinase 14
1.00
0.34
0.66
2.10
1.49
W61116


MAP kinase 8 interacting protein 2
2.90
0.16
0.35
0.24
0.55
AI202738


MAP kinase kinase kinase 12
1.48
0.20
0.91
0.58
0.68
AA053674


MAP kinase kinase kinase kinase 3
2.21
0.45
0.20
1.03
0.41
AA043537


MAP kinase kinase kinase 6
2.62
0.37
0.38

0.70
AW084649


MAP kinase kinase kinase kinase 4
1.04
0.96
0.09
0.29
2.79
AA417711


MAP kinase kinase kinase 11
1.53
0.65
0.41
0.99
0.44
R80779


MAP kinase kinase kinase 10
1.32
1.23
0.27
0.50
0.76
H01340


MAP kinase 9
2.54
0.57
0.39
0.16
0.38
AA157286


MAP kinase kinase kinase 1
1.23
0.61
0.42
0.81
1.07
AI538525


MAP kinase kinase kinase 8
0.66
1.52
1.82
9.50
0.59
W56266


MAP kinase-activated protein kinase 3
0.52
2.13
2.68
1.13
1.93
W68281


MAP kinase kinase 2
0.84
1.20
3.35
0.02
1.31
AA425826


MAP kinase kinase kinase 7
1.00
0.97

1.62
7.46
AA460969


MAP kinase 7
0.09

11.45
11.80
33.43
H39192


MAP kinase kinase 6
0.10
17.83
9.61

32.30
H07920


regulator of G-protein signaling 5
3.7397
0.27
0.06
0.68
0.18
AA668470


regulator of G-protein signaling 13
1.8564
0.54
0.45
0.07
1.09
H70047


G protein-coupled receptor
1.04
1.84
0.16
0.09
0.96
R91916


G protein-coupled receptor 17
1.78
0.32
0.56
0.39
0.77
AI953187


G protein-coupled receptor kinase 7
2.62

0.34
0.91
0.38
AA488413


orphan seven-transmembrane receptor,
7.16
1.06
0.10
0.11
0.14
AI131555


chemokine related


apoptosis antagonizing transcription
1.00
0.28
2.50
1.28
0.19
AI439571


factor


caspase 1, apoptosis-related cysteine
2.83
0.44

0.33
0.35
T95052


protease (interleukin 1, beta,


convertase)


programmed cell death 8 (apoptosis-
1.00
1.07
0.35
1.94
0.08
AA496348


inducing factor)







The cationic peptides at concentrations of 50 μg/ml were shown to decrease the expression of several polynucleotides. Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human cDNA arrays ID#PRHU03-S3. The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Unstimulated” columns refers to the intensity



# of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 23










Pro-inflammatory polynucleotides up-regulated by peptide treatment of A549


cells.











Unstim.
Ratio Peptide:Unstimulated
Accession













Polynucleotide/Protein and function
Intensity
ID 2
ID 3
ID 19
ID 1
Number
















IL-11 Rα; Receptor for pro-
0.55
2.39
0.98
4.85
1.82
AA454657


inflammatory cytokine, inflammation


IL-17 R; Receptor for IL-17, an inducer
0.54
2.05
1.97
1.52
1.86
AW029299


of cytokine production in epithelial cells


small inducible cytokine subfamily A,
1.00
3.88


2.41
AI922341


member 21; a chemokine


CD31; Leukocyte and cell to cell
0.59
2.71
3.13
1.01
1.68
R22412


adhesion (PECAM)


CCR6; Receptor for chemokine MIP-3α
0.14
4.51
7.75
6.92
7.79
N57964


integrin, alpha 2 (CD49B, alpha 2
1.00
0.89
2.44
3.62
0.88
AA463257


subunit of VLA-2 receptor; Adhesion to


leukocytes


integrin, alpha 3 (antigen CD49C, alpha
0.94
0.79
2.51
1.88
1.07
AA424695


3 subunit of VLA-3 receptor); Leukocyte


Adhesion


integrin, alpha E; Adhesion
0.01
179.33
120.12
28.48
81.37
AA425451


integrin, beta 4; Leukocyte adhesion
0.65
0.79
2.17
4.94
1.55
AA485668


C-type lectin-like receptor-2; Leukocyte
0.45
2.09
7.92
2.24
5.29
H70491


adhesion







The cationic peptides at concentrations of 50 μg/ml were shown to increase the expression of certain pro-inflammatory polynucleotides (data is a subset of Table 21). Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human cDNA arrays ID#PRHU03-S3. The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Unstimulated” columns



# refers to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 24










Pro-inflammatory polynucleotides down-regulated by peptide treatment of


A549 cells.











Unstim
Ratio Peptide:Unstimulated
Accession













Polynucleotide/Protein; Function
Intensity
ID 2
ID 3
ID 19
ID 1
Number
















Toll-like receptor (TLR) 1; Response to gram
3.22
0.35
0.31
0.14
0.19
AI339155


positive bacteria


TLR 2; Response to gram positive bacteria and
2.09
0.52
0.31
0.48
0.24
T57791


yeast


TLR 5; May augment other TLR responses,
8.01
0.12
0.39


N41021


Responsive to flagellin


TLR 7: Putative host defense mechanism
5.03
0.13
0.11
0.20
0.40
N30597


TNF receptor-associated factor 2; Inflammation
0.82
1.22
0.45
2.50
2.64
T55353


TNF receptor-associated factor 3; Inflammation
3.15
0.15

0.72
0.32
AA504259


TNF receptor superfamily, member 12;
4.17
0.59
0.24

0.02
W71984


Inflammation


TNF R superfamily, member 17; Inflammation
2.62

0.38
0.55
0.34
AA987627


TRAF and TNF receptor-associated protein;
1.33
0.75
0.22
0.67
0.80
AA488650


TNF signaling


small inducible cytokine subfamily A, member
2.26
0.32

0.44
1.26
AA495985


18; Chemokine


small inducible cytokine subfamily A, member
2.22
0.19
0.38
0.45
0.90
AI285199


20; Chemokine


small inducible cytokine subfamily A, member
2.64
0.38
0.31
1.53

AA916836


23; Chemokine


small inducible cytokine subfamily B, member 6
3.57
0.11
0.06
0.28
0.38
AI889554


(granulocyte chemotactic protein); Chemokine


small inducible cytokine subfamily B, member
2.02
0.50
1.07
0.29
0.40
AA878880


10; Chemokine


small inducible cytokine A3 (homologous to
2.84
1.79
0.32
0.35

AA677522


mouse Mip-1α); Chemokine


IL-12 receptor, beta 2; Interleukin and Interferon
4.58
0.67
0.22


AA977194


receptor


IL-18 receptor 1; Induces IFN-γ
1.78
0.50
0.42
0.92
0.56
AA482489


selectin L (lymphocyte adhesion molecule 1);
4.43
0.03
0.23
0.61

H00662


Leukocyte adhesion


vascular cell adhesion molecule 1; Leukocyte
1.40
0.20
0.72
0.77
0.40
H16591


adhesion


intercellular adhesion molecule 3; Leukocyte
1.00
0.12
0.31
2.04
1.57
AA479188


adhesion


integrin, alpha 1; Leukocyte adhesion
2.42
0.41
0.26

0.56
AA450324







The cationic peptides at concentrations of 50 μg/ml were shown to decrease the expression of certain pro-inflammatory polynucleotides (data is a subset of Table 22). Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human cDNA arrays ID#PRHU03-S3. The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Unstimulated” columns



# refers to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 25










Anti-inflammatory polynucleotides up-regulated by peptide treatment of A549


cells.











Unstim
Ratio Peptide:Unstimulated
Accession













Polynucleotide/Protein; Function
Intensity
ID 2
ID 3
ID 19
ID 1
Number
















IL-1 R antagonist homolog 1;
0.00
3085.96
1855.90
869.57

AI167887


Inhibitor of septic shock


IL-10 R beta; Receptor for
0.53
2.51
1.56
1.88
3.10
AA486393


cytokine synthesis inhibitor


TNF R, member 1B; Apoptosis
0.28
17.09
3.01
14.93
3.60
AA150416


TNF R, member 5; Apoptosis
33.71
2.98
0.02


H98636


(CD40L)


TNF R, member 11b; Apoptosis
1.00
5.29
4.50
0.78

AA194983


IK cytokine, down-regulator of
0.50
3.11
2.01
1.74
3.29
R39227


HLA II; Inhibits antigen


presentation


TGFB inducible early growth
0.90
2.38
2.08
0.87
1.11
AI473938


response 2; anti-inflammatory


cytokine


CD2; Adhesion molecule, binds
1.00
2.62
0.87
1.15
0.88
AA927710


LFAp3







The cationic peptides at concentrations of 50 μg/ml were shown to increase the expression of certain anti-inflammatory polynucleotides (data is a subset of Table 21). Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human cDNA arrays ID#PRHU03-S3. The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Unstimulated” columns



# refers to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 26










Anti-inflammatory polynucleotides down-regulated


by peptide treatment of A549 cells.










Polynucleotide/
Unstim
Ratio Peptide:Unstimulated
Accession













Protein; Function
Intensity
ID 2
ID 3
ID 19
ID 1
Number





MAP kinase 9
2.54
0.57
0.39
0.16
0.38
AA157286







The cationic peptides at concentrations of 50 μg/ml were shown to increase the expression of certain anti-inflammatory polynucleotides (data is a subset of Table 21). Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human cDNA arrays ID#PRHU03-S3. The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Unstimulated” columns



# refers to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 27










Polynucleotides up-regulated by SEQ ID NO: 6, in primary human macrophages.










Control:Unstimulated
Ratio peptide


Gene (Accession Number)
cells
treated:control












proteoglycan 2 (Z26248)
0.69
9.3


Unknown (AK001843)
26.3
8.2


phosphorylase kinase alpha 1 (X73874)
0.65
7.1


actinin, alpha 3 (M86407)
0.93
6.9


DKFZP586B2420 protein (AL050143)
0.84
5.9


Unknown (AL109678)
0.55
5.6


transcription factor 21 (AF047419)
0.55
5.4


Unknown (A433612)
0.62
5.0


chromosome condensation 1-like (AF060219)
0.69
4.8


Unknown (AL137715)
0.66
4.4


apoptosis inhibitor 4 (U75285)
0.55
4.2


TERF1 (TRF1)-interacting nuclear factor 2
0.73
4.2


(NM_012461)


LINE retrotransposable element 1 (M22333)
6.21
4.0


1-acylglycerol-3-phosphate O-acyltransferase 1
0.89
4.0


(U56417)


Vacuolar proton-ATPase, subunit D; V-
1.74
4.0


ATPase, subunit D (X71490)


KIAA0592 protein (AB011164)
0.70
4.0


potassium voltage-gated channel KQT-like
0.59
3.9


subfamily member 4 (AF105202)


CDC14 homolog A (AF000367)
0.87
3.8


histone fold proteinCHRAC17 (AF070640)
0.63
3.8


Cryptochrome 1 (D83702)
0.69
3.8


pancreatic zymogen granule membrane
0.71
3.7


associated protein (AB035541)


Sp3 transcription factor (X68560)
0.67
3.6


hypothetical protein FLJ20495 (AK000502)
0.67
3.5


E2F transcription factor 5, p130-binding
0.56
3.5


(U31556)


hypothetical protein FLJ20070 (AK000077)
1.35
3.4


glycoprotein IX (X52997)
0.68
3.4


KIAA1013 protein (AB023230)
0.80
3.4


eukaryotic translation initiation factor 4A,
2.02
3.4


isoform 2 (AL137681)


FYN-binding protein (AF198052)
1.04
3.3


guanine nucleotide binding protein, gamma
0.80
3.3


transducing activity polypeptide 1 (U41492)


glypican 1 (X54232)
0.74
3.2


mucosal vascular addressin cell adhesion
0.65
3.2


molecule 1 (U43628)


lymphocyte antigen (M38056)
0.70
3.2


H1 histone family, member 4 (M60748)
0.81
3.0


translational inhibitor protein p14.5 (X95384)
0.78
3.0


hypothetical protein FLJ20689 (AB032978)
1.03
2.9


KIAA1278 protein (AB03104)
0.80
2.9


unknown (AL031864)
0.95
2.9


chymotrypsin-like protease (X71877)
3.39
2.9


calumenin (NM_001219)
2.08
2.9


protein kinase, cAMP-dependent, regulatory,
7.16
2.9


type I, beta (M65066)


POU domain, class 4, transcription factor 2
0.79
2.8


(U06233)


POU domain, class 2, associating factor 1
1.09
2.8


(Z49194)


KIAA0532 protein (AB011104)
0.84
2.8


unknown (AF068289)
1.01
2.8


unknown (AL117643)
0.86
2.7


cathepsin E (M84424)
15.33
2.7


matrix metalloproteinase 23A (AF056200)
0.73
2.7


interferon receptor 2 (L42243)
0.70
2.5


MAP kinase kinase 1 (L11284)
0.61
2.4


protein kinase C, alpha (X52479)
0.76
2.4


c-Cbl-interacting protein (AF230904)
0.95
2.4


c-fos induced growth factor (Y12864)
0.67
2.3


cyclin-dependent kinase inhibitor 1B (S76988)
0.89
2.2


zinc finger protein 266 (X78924)
1.67
2.2


MAP kinase 14 (L35263)
1.21
2.2


KIAA0922 protein (AB023139)
0.96
2.1


bone morphogenetic protein 1 (NM_006129)
1.10
2.1


NADH dehydrogenase 1 alpha subcomplex, 10
1.47
2.1


(AF087661)


bone morphogenetic protein receptor, type IB
0.50
2.1


(U89326)


interferon regulatory factor 2 (NM 002199)
1.46
2.0


protease, serine, 21 (AB031331)
0.89
2.0







The peptide SEQ ID NO: 6 at a concentration of 50 μg/ml was shown to increase the expression of many polynucleotides. Peptide was incubated with the human macrophages for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio peptide



# treated:Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 28










Polynucleotides down-regulated by SEQ ID NO: 6, in primary human


macrophages.










Control:Unstimulated
Ratio peptide


Gene (Accession Number)
cells
treated:control












Unknown (AL049263)
17
0.06


integrin-linked kinase (U40282)
2.0
0.13


KIAA0842 protein (AB020649)
1.1
0.13


Unknown (AB037838)
13
0.14


Granulin (AF055008)
8.6
0.14


glutathione peroxidase 3 (NM_002084)
1.2
0.15


KIAA0152 gene product (D63486)
0.9
0.17


TGFB1-induced anti-apoptotic factor 1 (D86970)
0.9
0.19


disintegrin protease (Y13323)
1.5
0.21


proteasome subunit beta type 7 (D38048)
0.7
0.22


cofactor required for Sp1 transcriptional
0.9
0.23


activation subunit 3 (AB033042)


TNF receptor superfamily, member 14 (U81232)
0.8
0.26


proteasome 26S subunit non-ATPase 8 (D38047)
1.1
0.28


proteasome subunit beta type, 4 (D26600)
0.7
0.29


TNF receptor superfamily member 1B (M32315)
1.7
0.29


cytochrome c oxidase subunit Vic (X13238)
3.3
0.30


S100 calcium-binding protein A4 (M80563)
3.8
0.31


proteasome subunit alpha type, 6 (X59417)
2.9
0.31


proteasome 26S subunit non-ATPase, 10
1.0
0.32


(AL031177)


MAP kinase kinase kinase 2 (NM_006609)
0.8
0.32


ribosomal protein L11 (X79234)
5.5
0.32


matrix metalloproteinase 14 (Z48481)
1.0
0.32


proteasome subunit beta type, 5 (D29011)
1.5
0.33


MAP kinase-activated protein kinase 2 (U12779)
1.5
0.34


caspase 3 (U13737)
0.5
0.35


jun D proto-oncogene (X56681)
3.0
0.35


proteasome 26S subunit, ATPase, 3 (M34079)
1.3
0.35


IL-1 receptor-like 1 (AB012701)
0.7
0.35


interferon alpha-inducible protein (AB019565)
13
0.35


SDF receptor 1 (NM_012428)
1.6
0.35


Cathepsin D (M63138)
46
0.36


MAP kinase kinase 3 (D87116)
7.4
0.37


TGF, beta-induced, (M77349)
1.8
0.37


TNF receptor superfamily, member 10b
1.1
0.37


(AF016266)


proteasome subunit beta type, 6 (M34079)
1.3
0.38


nuclear receptor binding protein (NM_013392)
5.2
0.38


Unknown (AL050370)
1.3
0.38


protease inhibitor 1 alpha-1-antitrypsin (X01683)
0.7
0.40


proteasome subunit alpha type, 7 (AF054185)
5.6
0.40


LPS-induced TNF-alpha factor (NM_004862)
5.3
0.41


transferrin receptor (X01060)
14
0.42


proteasome 26S subunit non-ATPase 13
1.8
0.44


(AB009398)


MAP kinase kinase 5 (U25265)
1.3
0.44


Cathepsin L (X12451)
15
0.44


IL-1 receptor-associated kinase 1 (L76191)
1.7
0.45


MAP kinase kinase kinase kinase 2 (U07349)
1.1
0.46


peroxisome proliferative activated receptor delta
2.2
0.46


(AL022721)


TNF superfamily, member 15 (AF039390)
16
0.46


defender against cell death 1 (D15057)
3.9
0.46


TNF superfamily member 10 (U37518)
287
0.46


cathepsin H (X16832)
14
0.47


protease inhibitor 12 (Z81326)
0.6
0.48


proteasome subunit alpha type, 4 (D00763)
2.6
0.49


proteasome 26S subunit ATPase, 1 (L02426)
1.8
0.49


proteasome 26S subunit ATPase, 2 (D11094)
2.1
0.49


caspase 7 (U67319)
2.4
0.49


matrix metalloproteinase 7 (Z11887)
2.5
0.49







The peptide SEQ ID NO: 6 at a concentration of 50 μg/ml was shown to increase the expression of many polynucleotides. Peptide was incubated with the human macrophages for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio of Peptide:Control” columns refer to the intensity of



# polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 29










Polynucleotides up-regulated by SEQ ID NO: 1, in HBE cells.










Accession

Control:Unstimulated
Ratio peptide


Number
Gene
cells
treated:control













AL110161
Unknown
0.22
5218.3


AF131842
Unknown
0.01
573.1


AJ000730
solute carrier family
0.01
282.0


Z25884
chloride channel 1
0.01
256.2


M93426
protein tyrosine phosphatase receptor-
0.01
248.7



type, zeta


X65857
olfactory receptor, family 1, subfamily
0.01
228.7



D, member 2


M55654
TATA box binding protein
0.21
81.9


AK001411
hypothetical protein
0.19
56.1


D29643
dolichyl-diphosphooligosaccharide-
1.56
55.4



protein glycosyltransferase


AF006822
myelin transcription factor 2
0.07
55.3


AL117601
Unknown
0.05
53.8


AL117629
DKFZP434C245 protein
0.38
45.8


M59465
tumor necrosis factor, alpha-induced
0.50
45.1



protein 3


AB013456
aquaporin 8
0.06
41.3


AJ131244
SEC24 related gene family, member A
0.56
25.1


AL110179
Unknown
0.87
24.8


AB037844
Unknwon
1.47
20.6


Z47727
polymerase II polypeptide K
0.11
20.5


AL035694
Unknown
0.81
20.4


X68994

H. sapiens CREB gene

0.13
19.3


AJ238379
hypothetical protein
1.39
18.5


NM_003519
H2B histone family member
0.13
18.3


U16126
glutamate receptor, ionotropic kainate 2
0.13
17.9


U29926
adenosine monophosphate deaminase
0.16
16.3


AK001160
hypothetical protein
0.39
14.4


U18018
ets variant gene 4
0.21
12.9


D80006
KIAA0184 protein
0.21
12.6


AK000768
hypothetical protein
0.30
12.3


X99894
insulin promoter factor 1,
0.26
12.0


AL031177
Unknown
1.09
11.2


AF052091
unknown
0.28
10.9


L38928
5,10-methenyltetrahydrofolate
0.22
10.6



synthetase


AL117421
unknown
0.89
10.1


AL133606
hypothetical protein
0.89
9.8


NM_016227
membrane protein CH1
0.28
9.6


NM_006594
adaptor-related protein complex 4
0.39
9.3


U54996
ZW10 homolog, protein
0.59
9.3


AJ007557
potassium channel,
0.28
9.0


AF043938
muscle RAS oncogene
1.24
8.8


AK001607
unknown
2.74
8.7


AL031320
peroxisomal biogenesis factor 3
0.31
8.4


D38024
unknown
0.31
8.3


AF059575
LIM homeobox TF
2.08
8.2


AF043724
hepatitis A virus cellular receptor 1
0.39
8.1


AK002062
hypothetical protein
2.03
8.0


L13436
natriuretic peptide receptor
0.53
7.8


U33749
thyroid transcription factor 1
0.36
7.6


AF011792
cell cycle progression 2 protein
0.31
7.6


AK000193
hypothetical protein
1.18
6.8


AF039022
exportin, tRNA
0.35
6.8


M17017
interleukin 8
0.50
6.7


AF044958
NADH dehydrogenase
0.97
6.5


U35246
vacuolar protein sorting
0.48
6.5


AK001326
tetraspan 3
1.59
6.5


M55422
Krueppel-related zinc finger protein
0.34
6.4


U44772
palmitoyl-protein thioesterase
1.17
6.3


AL117485
hypothetical protein
0.67
5.9


AB037776
unknown
0.75
5.7


AF131827
unknown
0.69
5.6


AL137560
unknown
0.48
5.2


X05908
annexin A1
0.81
5.1


X68264
melanoma adhesion molecule
0.64
5.0


AL161995
neurturin
0.86
4.9


AF037372
cytochrome c oxidase
0.48
4.8


NM_016187
bridging integrator 2
0.65
4.8


AL137758
unknown
0.57
4.8


U59863
TRAF family member-associated NFKB
0.46
4.7



activator


Z30643
chloride channel Ka
0.70
4.7


D16294
acetyl-Coenzyme A acyltransferase 2
1.07
4.6


AJ132592
zinc finger protein 281
0.55
4.6


X82324
POU domain TF
1.73
4.5


NM_016047
CGI-110 protein
1.95
4.5


AK001371
hypothetical protein
0.49
4.5


M60746
H3 histone family member D
3.05
4.5


AB033071
hypothetical protein
4.47
4.4


AB002305
KJAA0307 gene product
1.37
4.4


X92689
UDP-N-acetyl-alpha-D-
0.99
4.4



galactosamine:polypeptide N-



acetylgalactosaminyltransferase 3


AL049543
glutathione peroxidase 5
1.62
4.3


U43148
patched homolog
0.96
4.3


M67439
dopamine receptor D5
2.61
4.2


U09850
zinc finger protein 143
0.56
4.2


L20316
glucagon receptor
0.75
4.2


AB037767
a disintegrin-like and metalloprotease
0.69
4.2


NM_017433
myosin IIIA
99.20
4.2


D26579
a disintegrin and metalloprotease domain 8
0.59
4.1


L10333
reticulon 1
1.81
4.1


AK000761
unknown
1.87
4.1


U91540
NK homeobox family 3, A
0.80
4.1


Z17227
interleukin 10 receptor, beta
0.75
4.0







The peptide SEQ ID NO: 1 at a concentration of 50 μg/ml was shown to increase the expression of many polynucleotides. Peptide was incubated with the human HBE epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in unstimulated cells is shown in the second column. The “Ratio Peptide:Control” columns refer to the intensity of



# polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 30










Polynucleotides down-regulated by Peptide (50 μg/ml), SEQ ID NO: 1, in HBE cells.













Ratio of SEQ ID


Accession

Control:Unstimulated
NO: 1-


Number
Gene
Cells
treated:control













AC004908
Unknown
32.4
0.09


S70622
G1 phase-specific gene
43.1
0.10


Z97056
DEAD/H box polypeptide
12.8
0.11


AK002056
hypothetical protein
11.4
0.12


L33930
CD24 antigen
28.7
0.13


X77584
thioredoxin
11.7
0.13


NM_014106
PRO1914 protein
25.0
0.14


M37583
H2A histone family member
22.2
0.14


U89387
polymerase (RNA) II polypeptide D
10.2
0.14


D25274
ras-related C3 botulinum toxin substrate 1
10.3
0.15


J04173
phosphoglycerate mutase 1
11.4
0.15


U19765
zinc finger protein 9
8.9
0.16


X67951
proliferation-associated gene A
14.1
0.16


AL096719
profilin 2
20.0
0.16


AF165217
tropomodulin 4
14.6
0.16


NM_014341
mitochondrial carrier homolog 1
11.1
0.16


AL022068
Unknown
73.6
0.17


X69150
ribosomal protein S18
42.8
0.17


AL031577
Unknown
35.0
0.17


AL031281
Unknown
8.9
0.17


AF090094
Human mRNA for ornithine decarboxylase
10.3
0.17



antizyme,


AL022723
HLA-G histocompatibility antigen, class I, G
20.6
0.18


U09813
ATP synthase, H+ transporting mitochondrial
9.8
0.18



F0 complex


AF000560
Homo sapiens TTF-I interacting peptide 20
20.2
0.19


NM_016094
HSPC042 protein
67.2
0.19


AF047183
NADH dehydrogenase
7.5
0.19


D14662
anti-oxidant protein 2 (non-selenium
8.1
0.19



glutathione peroxidase, acidic calcium-



independent phospholipas


X16662
annexin A8
8.5
0.19


U14588
paxillin
11.3
0.19


AL117654
DKFZP586D0624 protein
12.6
0.20


AK001962
hypothetical protein
7.7
0.20


L41559
6-pyruvoyl-tetrahydropterin
9.1
0.20



synthase/dimerization cofactor of hepatocyte



nuclear factor 1 alpha


NM_016139
16.7 Kd protein
21.0
0.21


NM_016080
CGI-150 protein
10.7
0.21


U86782
26S proteasome-associated pad 1 homolog
6.7
0.21


AJ400717
tumor protein, translationally-controlled 1
9.8
0.21


X07495
homeo box C4
31.0
0.21


AL034410
Unknown
7.3
0.22


X14787
thrombospondin 1
26.2
0.22


AF081192
purine-rich element binding protein B
6.8
0.22


D49489
protein disulfude isomerase-related protein
11.0
0.22


NM_014051
PTD011 protein
9.3
0.22


AK001536
Unknown
98.0
0.22


X62534
high-mobility group protein 2
9.5
0.22


AJ005259
endothelial differentiation-related factor 1
6.7
0.22


NM_000120
epoxide hydrolase 1, microsomal
10.0
0.22


M38591
S100 calcium-binding protein A10
23.9
0.23


AF071596
immediate early response 3
11.5
0.23


X16396
methylene tetrahydrofolate dehydrogenase
8.3
0.23


AK000934
ATPase inhibitor precursor
7.6
0.23


AL117612
Unknown
10.7
0.23


AF119043
transcriptional intermediary factor 1 gamma
7.3
0.23


AF037066
solute carrier family 22 member 1-like
7.6
0.23



antisense


AF134406
cytochrome c oxidase subunit
13.3
0.23


AE000661
Unknown
9.2
0.24


AL157424
synaptojanin 2
7.2
0.24


X56468
tyrosine 3-monooxygenase/tryptophan 5-
7.2
0.24



monooxygenase activation protein,


U39318
ubiquitin-conjugating enzyme E2D 3
10.7
0.24


AL034348
Unknown
24.4
0.24


D26600
proteasome subunit beta type 4
11.4
0.24


AB032987
Unknown
16.7
0.24


J04182
lysosomal-associated membrane protein 1
7.4
0.24


X78925
zinc finger protein 267
16.1
0.25


NM_000805
gastrin
38.1
0.25


U29700
anti-Mullerian hormone receptor, type II
12.0
0.25


Z98200
Unknown
13.4
0.25


U07857
signal recognition particle
10.3
0.25


L05096
Homo sapiens ribosomal protein L39
25.3
0.25


AK001443
hypothetical protein
7.5
0.25


K03515
glucose phosphate isomerase
6.2
0.25


X57352
interferon induced transmembrane protein 3
7.5
0.26


J02883
colipase pancreatic
5.7
0.26


M24069
cold shock domain protein
6.3
0.26


AJ269537
chondroitin-4-sulfotransferase
60.5
0.26


AL137555
Unknown
8.5
0.26


U89505
RNA binding motif protein 4
5.5
0.26


U82938
CD27-binding protein
7.5
0.26


X99584
SMT3 homolog 1
12.8
0.26


AK000847
Unknown
35.8
0.27


NM_014463
Lsm3 protein
7.8
0.27


AL133645
Unknown
50.8
0.27


X78924
zinc finger protein 266
13.6
0.27


NM_004304
anaplastic lymphoma kinase
15.0
0.27


X57958
ribosomal protein L7
27.9
0.27


U63542
Unknown
12.3
0.27


AK000086
hypothetical protein
8.3
0.27


X57138
H2A histone family member N
32.0
0.27


AB023206
KIAA0989 protein
6.5
0.27


AB021641
gonadotropin inducible transcriptn repressor-1,
5.5
0.28


AF050639
NADH dehydrogenase
5.5
0.28


M62505
complement component 5 receptor 1
7.5
0.28


X64364
basigin
5.8
0.28


AJ224082
Unknown
22.5
0.28


AF042165
cytochrome c oxidase
20.4
0.28


AK001472
anillin
10.9
0.28


X86428
protein phosphatase 2A subunit
12.7
0.28


AF227132
candidate taste receptor T2R5
5.1
0.28


Z98751
Unknown
5.3
0.28


D21260
clathrin heavy polypeptide
8.3
0.28


AF041474
actin-like 6
15.1
0.28


NM_005258
GTP cyclohydrolase I protein
7.6
0.28


L20859
solute carrier family 20
9.6
0.29


Z80783
H2B histone family member
9.0
0.29


AB011105
laminin alpha 5
7.1
0.29


AL008726
protective protein for beta-galactosidase
5.2
0.29


D29012
proteasome subunit
12.6
0.29


X63629
cadherin 3 P-cadherin
6.8
0.29


X02419
plasminogen activator urokinase
12.9
0.29


X13238
cytochrome c oxidase
8.0
0.29


X59798
cyclin D1
12.7
0.30


D78151
proteasome 26S subunit
7.6
0.31


AF054185
proteasome subunit
18.8
0.31


J03890
surfactant pulmonary-associated protein C
5.5
0.32


M34079
proteasome 26S subunit,
5.2
0.33







The peptide SEQ ID NO: 1 at a concentration of 50 μg/ml was shown to decrease the expression of many polynucleotides. Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in unstimulated cells is shown in the third column. The “Ratio Peptide:Control” columns refer to the intensity of



# polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 31










Up-regulation of Polynucleotide expression in A549 cells induced by Formula A


Peptides.
















Accession

ctrl-
ctrl I-








Number
Gene
Cy3
Cy5
ID 5:ctrl
ID 6:ctrl
ID 7:ctrl
ID 8:ctrl
ID 9:ctrl
ID 10:ctrl



















U12472
glutathione S-
0.09
0.31
13.0
3.5
4.5
7.0
4.3
16.4



transferase


X66403
cholinergic
0.17
0.19
7.8
9.9
6.0
6.4
5.0
15.7



receptor


AK001932
unknown
0.11
0.25
19.4
4.6
9.9
7.6
8.1
14.5


X58079
S100 calcium-
0.14
0.24
12.2
7.6
8.1
4.3
4.5
13.2



binding



protein


U18244
solute carrier
0.19
0.20
6.1
9.7
11.9
5.0
3.7
10.6



family 1


U20648
zinc finger
0.16
0.13
5.3
6.2
5.6
3.1
6.8
9.5



protein


AB037832
unknown
0.10
0.29
9.0
4.2
9.4
3.1
2.6
8.7


AC002542
unknown
0.15
0.07
10.5
15.7
7.8
10.1
11.7
8.2


M89796
membrane-
0.15
0.14
2.6
6.1
7.6
3.5
13.3
8.1



spanning 4-



domains,



subfamily A


AF042163
cytochrome c
0.09
0.19
3.9
3.2
7.6
6.3
4.9
7.9



oxidase


AL032821
Vanin 2
0.41
0.23
2.5
5.2
3.2
2.1
4.0
7.9


U25341
melatonin
0.04
0.24
33.1
5.1
23.3
6.6
4.1
7.6



receptor 1B


U52219
G protein-
0.28
0.20
2.1
6.2
6.9
2.4
3.9
7.1



coupled



receptor


X04506
apolipoprotein B
0.29
0.32
7.9
3.4
3.3
4.8
2.6
7.0


AB011138
ATPase type
0.12
0.07
3.5
12.9
6.6
6.4
21.3
6.9



IV


AF055018
unknown
0.28
0.22
3.8
6.9
5.0
2.3
3.1
6.8


AK002037
hypothetical
0.08
0.08
2.9
7.9
14.1
7.9
20.1
6.5



protein


AK001024
guanine
0.16
0.11
7.7
11.9
5.0
10.3
6.0
6.3



nucleotide-



binding



protein


AF240467
TLR-7
0.11
0.10
20.4
9.0
3.4
9.4
12.9
6.1


AF105367
glucagon-like
0.15
0.35
23.2
2.6
3.0
10.6
2.9
5.7



peptide 2



receptor


AL009183
TNFR
0.46
0.19
10.6
4.7
3.7
2.8
6.5
5.7



superfamily,



member 9


X54380
pregnancy-
0.23
0.08
4.7
11.9
7.2
12.7
3.8
5.5



zone protein


AL137736
unknown
0.22
0.15
2.1
7.2
3.3
7.1
4.6
5.5


X05615
thyroglobulin
0.28
0.42
6.3
2.7
7.7
2.4
3.1
5.4


D28114
myelin-
0.24
0.08
2.5
15.9
13.0
7.1
13.7
5.4



associated



protein


AK000358
microfibrillar-
0.28
0.28
8.7
4.2
7.2
3.2
2.4
5.3



associated



protein 3


AK001351
unknown
0.12
0.22
3.9
7.6
8.7
3.9
2.3
5.2


U79289
unknown
0.14
0.27
2.5
2.7
2.8
2.0
4.3
5.1


AB014546
ring finger
0.12
0.34
6.8
2.4
4.1
2.7
2.0
5.0



protein


AL117428
DKFZP434A2
0.10
0.07
2.8
16.1
12.8
9.7
14.2
4.9



36 protein


AL050378
unknown
0.41
0.14
3.5
8.7
11.7
3.5
7.0
4.9


AJ250562
transmembrane
0.13
0.10
5.2
5.7
14.2
3.8
10.3
4.8



4 superfamily



member 2


NM_001756
corticosteroid
0.28
0.13
4.0
7.9
6.5
14.9
5.6
4.8



binding



globulin


AL137471
hypothetical
0.29
0.05
3.7
18.0
6.2
7.2
16.3
4.7



protein


M19684
protease
0.41
0.14
3.5
4.6
5.4
2.8
9.4
4.7



inhibitor 1


NM_001963
epidermal
0.57
0.05
3.4
6.2
1.8
32.9
14.7
4.4



growth factor


NM_000910
neuropeptide
0.62
0.36
3.1
2.7
2.3
2.6
3.1
4.4



Y receptor


AF022212
Rho GTPase
0.19
0.02
9.0
45.7
25.6
12.4
72.2
4.4



activating



protein 6


AK001674
cofactor
0.11
0.13
8.4
6.5
7.9
4.5
7.4
4.3



required for



Sp1


U51920
signal
0.23
0.27
3.4
3.8
2.1
4.1
8.8
4.2



recognition



particle


AK000576
hypothetical
0.27
0.06
4.4
14.7
7.4
14.1
8.6
4.2



protein


AL080073
unknown
0.17
0.20
21.6
3.9
4.3
8.8
2.6
4.1


U59628
paired box
0.34
0.06
3.4
14.1
5.4
7.9
4.9
4.1



gene 9


U90548
butyrophilin,
0.41
0.31
2.3
4.7
5.5
6.8
3.4
4.1



subfamily 3,



member A3


M19673
cystatin SA
0.43
0.26
2.3
8.5
4.5
2.5
4.1
3.8


AL161972
ICAM 2
0.44
0.37
2.0
3.6
2.0
2.7
5.5
3.8


X54938
inositol 1,4,5-
0.32
0.22
3.9
3.3
6.2
3.1
4.4
3.7



trisphosphate



3-kinase A


AB014575
KIAA0675
0.04
0.13
46.2
4.5
10.2
8.0
6.2
3.4



gene product


M83664
MHC II, DP
0.57
0.29
2.9
2.1
2.0
3.1
6.6
3.4



beta 1


AK000043
hypothetical
0.34
0.14
2.7
7.1
3.7
9.4
8.8
3.3



protein


U60666
testis specific
0.21
0.11
9.9
9.0
4.1
5.5
13.0
3.3



leucine rich



repeat protein


AK000337
hypothetical
0.49
0.19
4.3
5.1
4.7
10.6
7.1
3.3



protein


AF050198
putative
0.34
0.15
7.0
6.3
3.6
5.6
11.9
3.3



mitochondrial



space protein


AJ251029
odorant-
0.28
0.12
4.4
9.4
7.2
8.8
7.1
3.2



binding



protein 2A


X74142
forkhead box
0.12
0.33
19.5
4.5
8.4
6.4
4.4
3.2



G1B


AB029033
KIAA1110
0.35
0.24
3.1
2.2
5.6
5.2
3.1
3.1



protein


D85606
cholecystokinin
0.51
0.14
4.3
3.9
4.6
3.5
7.2
3.1



A receptor


X84195
acylphosphatase
0.32
0.19
4.8
3.7
5.0
11.2
9.8
3.0



2 muscle type


U57971
ATPase Ca++
0.29
0.13
2.2
7.9
1.8
6.3
4.8
3.0



transporting



plasma



membrane 3


J02611
apolipoprotein D
0.28
0.10
2.8
11.0
3.7
10.3
8.4
3.0


AF071510
lecithin retinol
0.07
0.05
7.9
3.8
11.7
46.0
16.3
3.0



acyltransferase


AF131757
unknown
0.10
0.08
4.8
9.0
44.3
9.3
10.7
3.0


L10717
IL2-inducible
0.45
0.21
2.5
4.9
2.8
10.9
4.5
2.9



T-cell kinase


L32961
4-aminobutyrate
0.64
0.32
3.6
2.9
3.2
5.3
2.3
2.9



aminotransferase


NM_003631
poly (ADP-
0.46
0.41
9.7
3.9
4.1
3.8
2.8
2.7



ribose)



glycohydrolase


AF098484
pronapsin A
0.28
0.14
3.7
3.7
5.6
11.6
3.7
2.5


NM_009589
arylsulfatase D
0.73
0.16
3.2
5.6
6.0
48.6
7.2
2.4


M14764
TNFR
0.49
0.15
2.3
3.5
10.6
13.6
6.8
2.2



superfamily,



member 16


AL035250
endothelin 3
0.52
0.14
2.1
7.3
4.8
4.5
3.7
2.2


M97925
defensin,
0.33
0.07
4.0
14.7
7.8
9.4
3.5
2.1



alpha 5,



Paneth cell-



specific


D43945
transcription
0.46
0.19
6.6
2.9
8.2
4.0
3.5
2.1



factor EC


D16583
histidine
0.46
0.09
3.2
13.8
4.2
8.8
13.7
2.1



decarboxylase







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides. Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labeling of cDNA with the dyes Cy3 and Cy5 respectively.



# The “ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 32










Up-regulation of Polynucleotide expression in A549 cells induced by Formula B


Peptides.
















Accession

ctrl-
ctrl-








Number
Gene
Cy3
Cy5
ID 12:ctrl
ID 13:ctrl
ID 14:ctrl
ID 15:ctrl
ID 16:ctrl
ID 17:ctrl



















AL157466
unknown
0.05
0.06
18.0
21.4
16.7
5.2
6.8
8.6


AB023215
KIAA0998
0.19
0.07
14.8
10.6
7.9
14.4
6.6
16.1



protein


AL031121
unknown
0.24
0.09
14.1
5.7
3.8
5.5
2.8
4.6


NM_016331
zinc finger
0.16
0.08
12.8
7.2
11.0
5.3
11.2
9.7



protein


M14565
cytochrome
0.16
0.12
10.6
12.5
5.0
3.6
10.1
6.3



P450


U22492
G protein-
0.28
0.07
10.4
8.9
4.8
10.8
6.6
3.6



coupled



receptor 8


U76010
solute carrier
0.14
0.07
9.7
18.6
3.7
4.8
5.6
8.9



family 30


AK000685
unknown
0.51
0.10
9.0
3.1
2.8
3.9
15.3
3.0


AF013620
Immunoglobulin
0.19
0.18
8.5
2.6
6.2
5.7
8.2
3.8



heavy variable



4-4


AL049296
unknown
0.61
0.89
8.1
3.2
2.7
3.2
2.7
2.0


AB006622
KIAA0284
0.47
0.28
7.5
5.0
2.8
11.1
5.5
4.6



protein


X04391
CD5 antigen
0.22
0.13
7.2
16.7
2.7
7.7
6.1
5.9


AK000067
hypothetical
0.80
0.35
7.1
4.6
2.1
3.2
8.5
2.2



protein


AF053712
TNF
0.17
0.08
6.9
17.7
3.0
6.2
12.3
5.2



superfamily_member



11


X58079
S100 calcium-
0.14
0.24
6.7
6.7
5.9
6.5
5.3
2.5



binding protein



A1


M91036
hemoglobin_gamma A
0.48
0.36
6.7
14.2
2.1
2.9
2.7
4.8


AF055018
unknown
0.28
0.22
6.3
10.7
2.7
2.6
4.6
6.5


L17325
pre-T/NK cell
0.19
0.29
6.1
4.4
6.5
4.7
4.0
4.0



associated



protein


D45399
phosphodiesterase
0.21
0.18
6.1
4.6
5.0
2.8
10.8
4.0


AB023188
KIAA0971
0.29
0.13
5.9
10.6
3.6
3.4
10.6
7.2



protein


NM_012177
F-box protein
0.26
0.31
5.9
5.5
3.8
2.8
3.0
6.8


D38550
E2F TF 3
0.43
0.39
5.8
3.4
2.1
4.5
2.5
2.4


AL050219
unknown
0.26
0.04
5.7
17.0
3.1
9.2
30.3
16.1


AL137540
unknown
0.67
0.79
5.5
3.2
3.9
10.9
2.9
2.3


D50926
KIAA0136
0.57
0.21
5.4
5.6
2.0
3.3
4.4
3.2



protein


AL137658
unknown
0.31
0.07
5.4
12.1
2.6
10.8
3.9
8.6


U21931
fructose-
0.48
0.14
5.4
4.1
2.9
3.6
6.0
3.2



bisphosphatase 1


AK001230
DKFZP586D21
0.43
0.26
5.0
4.6
2.1
2.2
2.5
2.7



1 protein


AL137728
unknown
0.67
0.47
5.0
5.9
2.2
6.8
5.9
2.1


AB022847
unknown
0.39
0.24
4.5
2.2
3.5
4.3
3.8
3.7


X75311
mevalonate
0.67
0.22
4.3
4.0
2.0
8.3
4.0
5.1



kinase


AK000946
DKFZP566C24
0.36
0.29
4.1
3.8
3.9
5.4
25.8
2.7



3 protein


AB023197
KIAA0980
0.25
0.30
4.0
8.3
2.1
8.8
2.2
4.9



protein


AB014615
fibroblast
0.19
0.07
3.9
3.3
7.0
3.4
2.2
7.7



growth factor 8


X04014
unknown
0.29
0.16
3.8
2.5
2.2
3.0
5.5
3.1


U76368
solute carrier
0.46
0.17
3.8
3.8
2.8
3.2
4.2
3.0



family 7


AB032436
unknown
0.14
0.21
3.8
2.7
6.1
3.2
4.5
2.6


AB020683
KIAA0876
0.37
0.21
3.7
4.2
2.2
5.3
2.9
9.4



protein


NM_012126
carbohydrate
0.31
0.20
3.7
5.2
3.2
3.4
3.9
2.5



sulfotransferase 5


AK002037
hypothetical
0.08
0.08
3.7
17.1
4.6
12.3
11.0
8.7



protein


X78712
glycerol kinase
0.17
0.19
3.6
2.5
4.5
5.3
2.2
3.3



pseudogene 2


NM_014178
HSPC156
0.23
0.12
3.5
8.4
2.9
6.9
14.4
5.5



protein


AC004079
homeo box A2
0.31
0.11
3.5
7.0
2.1
2.0
7.3
9.1


AL080182
unknown
0.51
0.21
3.4
3.5
2.2
2.1
2.9
2.4


M91036
hemoglobin
0.22
0.02
3.4
26.3
5.8
6.8
30.4
21.6



gamma G


AJ000512
serum/glucocorticoid
0.27
0.43
3.3
2.1
4.9
2.3
3.9
2.7



regulated



kinase


AK002140
hypothetical
0.28
0.14
3.3
9.9
2.8
2.1
16.6
7.2



protein


AL137284
unknown
0.22
0.04
3.3
7.2
4.1
6.0
12.2
3.7


Z11898
POU domain_class
0.12
0.29
3.2
3.7
8.2
2.5
6.6
2.2



5 TF 1


AB017016
brain-specific
0.27
0.29
3.1
2.8
2.5
2.8
3.3
5.5



protein


X54673
Solute-carrier
0.34
0.08
2.9
12.0
2.2
10.4
7.4
5.9



family 6


AL033377
unknown
0.40
0.22
2.6
2.6
2.6
2.3
4.5
2.2


X85740
CCR4
0.34
0.05
2.6
2.3
2.6
2.5
12.5
5.2


AB010419
core-binding
0.59
0.20
2.5
12.8
2.0
2.8
2.9
5.9



factor


AL109726
uknown
0.14
0.15
2.3
9.0
4.3
4.4
2.6
3.7


NM_012450
sulfate
0.15
0.10
2.2
3.1
8.2
9.9
4.7
5.9



transporter 1


J04599
biglycan
0.39
0.30
2.1
3.3
6.6
2.2
2.7
5.4


AK000266
hypothetical
0.49
0.35
2.1
3.5
3.5
6.6
4.3
4.0



protein







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides. Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labeling of cDNA with the dyes Cy3 and Cy5 respectively.



# The “ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 33










Up-regulation of Polynucleotide expression in A549 cells induced by Formula C


Peptides.
















Accession

ctrl-
ctrl-








Number
Gene
Cy3
Cy5
ID 19:ctrl
ID 20:ctrl
ID 21:ctrl
ID 22:ctrl
ID 23:ctrl
ID 24:ctrl



















NM_014139
sodium
0.04
0.05
31.6
25.2
18.0
9.7
22.2
11.2



channel



voltage-



gated,


X84003
TATA box
0.47
0.07
31.8
12.7
2.5
2.8
18.0
14.2



binding



protein


AF144412
lens epithelial
0.25
0.07
23.9
8.0
6.8
3.4
16.2
3.5



cell protein


AL080107
unknown
0.11
0.06
17.8
34.4
12.4
6.2
5.4
7.9


AF052116
unknown
0.34
0.07
15.5
3.9
9.2
3.0
6.9
2.7


AB033063
unknown
0.46
0.13
15.2
10.3
4.0
2.6
7.2
11.2


AK000258
hypothetical
0.27
0.07
13.9
8.0
3.5
3.4
26.5
11.5



protein


NM_006963
zinc finger
0.10
0.08
12.8
6.8
6.2
5.9
17.2
1241.2



protein


NM_014099
PRO1768
0.30
0.06
12.3
17.4
5.4
5.4
19.5
3.4



protein


AK000996
hypothetical
0.17
0.07
10.0
8.0
9.7
7.4
20.7
16.3



protein


M81933
cell division
0.13
0.21
8.8
7.8
19.6
15.6
4.8
3.8



cycle 25A


AF181286
unknown
0.05
0.22
8.8
2.7
12.0
35.6
5.9
2.3


AJ272208
IL-1R
0.22
0.17
8.8
2.9
5.0
3.2
9.8
7.3



accessory



protein-like 2


AF030555
fatty-acid-
0.10
0.39
8.7
2.2
11.3
9.9
3.0
2.1



Coenzyme A



ligase


AL050125
unknown
0.23
0.07
8.6
14.3
5.2
2.8
18.7
8.3


AB011096
KIAA0524
0.21
0.08
8.5
24.4
4.7
6.8
10.4
7.5



protein


J03068
N-
0.54
0.21
8.3
2.4
2.2
4.1
3.0
6.0



acylaminoacyl-



peptide



hydrolase


M33906
MHC class
0.14
0.08
7.6
4.5
15.2
6.1
7.5
7.9



II, DQ alpha 1


AJ272265
secreted
0.21
0.09
7.6
9.0
3.3
4.9
18.8
14.5



phosphoprotein


J00210
interferon
0.41
0.07
7.2
15.0
2.8
3.1
11.0
4.3



alpha 13


AK001952
hypothetical
0.42
0.21
6.9
4.9
2.5
3.1
7.6
4.5



protein


X54131
protein
0.09
0.20
6.4
6.5
7.7
15.0
5.6
4.1



tyrosine



phosphatase,



receptor type,


AF064493
LIM binding
0.46
0.14
5.9
5.6
2.2
2.9
8.5
5.8



domain 2


AL117567
DKFZP566O
0.44
0.22
5.8
3.3
2.9
2.3
5.7
14.9



084 protein


L40933
phosphogluco
0.16
0.03
5.6
11.0
4.8
3.5
8.5
76.3



mutase 5


M27190
regenerating
0.19
0.28
5.3
3.0
3.8
3.6
5.8
3.6



islet-derived



1 alpha


AL031121
unknown
0.24
0.09
5.3
3.8
3.2
3.9
3.0
27.9


U27655
regulator of
0.24
0.29
5.0
9.0
4.5
8.3
4.2
4.5



G-protein



signaling


AB037786
unknown
0.12
0.03
4.7
54.1
2.8
2.3
2.2
11.0


X73113
myosin-
0.29
0.13
4.7
6.5
6.0
2.4
6.7
6.3



binding



protein C


AB010962
matrix
0.08
0.12
4.7
6.2
2.4
4.7
10.9
4.2



metalloproteinase


AL096729
unknown
0.36
0.13
4.7
7.7
3.2
2.4
6.3
6.2


AB018320
Arg/Abl-
0.16
0.18
4.6
7.1
3.0
3.3
5.8
8.9



interacting



protein


AK001024
guanine
0.16
0.11
4.6
2.0
9.8
2.6
7.6
14.1



nucleotide-



binding



protein


AJ275355
unknown
0.15
0.08
4.6
17.3
5.4
9.2
5.1
5.5


U21931
fructose-
0.48
0.14
4.6
4.3
2.6
2.1
8.4
9.6



bisphosphatase 1


X66403
cholinergic
0.17
0.19
4.4
9.0
10.9
9.3
5.1
6.7



receptor


X67734
contactin 2
0.25
0.09
4.3
6.8
3.1
5.8
7.9
8.4


U92981
unknown
0.20
0.23
4.3
3.2
4.8
5.6
5.4
6.3


X68879
empty
0.05
0.08
4.3
2.0
12.3
2.7
5.6
4.7



spiracles



homolog 1


AL137362
unknown
0.22
0.22
4.2
4.1
2.7
4.1
9.3
4.2


NM_001756
corticosteroid
0.28
0.13
4.1
10.6
3.9
2.7
10.3
5.5



binding



globulin


U80770
unknown
0.31
0.14
4.1
4.1
23.3
2.7
7.0
10.1


AL109792
unknown
0.16
0.19
4.0
4.5
4.3
8.8
8.7
3.9


X65962
cytochrome
0.33
0.05
3.8
25.3
5.7
5.1
19.8
12.0



P-450


AK001856
unknown
0.40
0.21
3.8
7.0
2.6
3.1
2.9
7.8


AL022723
MHC, class I, F
0.55
0.18
3.7
5.7
4.4
2.3
3.3
5.2


D38449
putative G
0.18
0.09
3.5
11.1
13.3
5.8
4.8
5.2



protein



coupled



receptor


AL137489
unknown
0.74
0.26
3.3
2.9
2.6
3.3
2.5
5.4


AB000887
small
0.76
0.18
3.3
5.0
2.6
2.4
5.9
10.3



inducible



cytokine



subfamily A


NM_012450
sulfate
0.15
0.10
3.3
9.0
10.0
10.9
4.6
8.7



transport 1


U86529
glutathione
0.55
0.15
3.2
6.8
4.4
2.3
9.3
5.1



S-transferase



zeta 1


AK001244
unknown
0.79
0.31
3.2
5.5
2.3
2.3
3.9
2.8


AL133602
unknown
0.16
0.21
3.1
7.8
8.7
2.6
4.1
5.6


AB033080
cell cycle
0.31
0.31
3.1
4.6
3.0
3.5
2.2
4.2



progression 8



protein


AF023466
putative
0.27
0.18
3.1
5.0
4.2
7.4
10.1
3.8



glycine-N-



acyltransferase


AL117457
cofilin 2
0.68
0.53
3.0
4.6
3.3
2.4
7.4
3.4


AC007059
unknown
0.37
0.35
3.0
5.7
3.1
2.4
2.6
2.4


U60179
growth
0.34
0.21
2.9
3.5
2.3
3.1
8.0
4.7



hormone



receptor


M37238
phospholipase
0.60
0.36
2.9
2.0
3.2
2.1
2.9
4.6



C, gamma 2


L22569
cathepsin B
0.32
0.12
2.9
2.1
6.2
3.0
13.1
16.7


M80359
MAP/microtubule
0.37
0.76
2.9
3.1
6.1
7.6
2.1
3.3



affinity-



regulating



kinase 3


S70348
Integrin beta 3
0.58
0.31
2.6
4.8
4.1
2.6
2.6
2.6


L13720
growth
0.36
0.26
2.4
2.5
6.8
4.8
3.9
3.7



arrest-



specific 6


AL049423
unknown
0.33
0.30
2.4
3.7
3.8
2.8
2.9
3.4


AL050201
unknown
0.68
0.29
2.2
3.1
3.7
3.0
3.0
2.2


AF050078
growth arrest
0.87
0.33
2.1
8.4
2.5
2.2
2.6
4.4



specific 11


AK001753
hypothetical
0.53
0.28
2.1
5.0
2.2
2.8
3.6
4.6



protein


X05323
unknown
0.39
0.13
2.1
7.8
2.6
2.4
21.5
3.5


AB014548
KIAA0648
0.61
0.30
2.0
2.4
4.8
3.4
4.9
3.9



protein







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides. Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04). The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labeling of cDNA with the dyes Cy3 and Cy5 respectively.



# The “ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.













TABLE 34










Up-regulation of Polynucleotide expression in A549 cells induced by Formula D


Peptides.
















Accession

ctrl-
ctrl-








Number
Gene
Cy3
Cy5
ID 26:ctrl
ID 27:ctrl
ID 28:ctrl
ID 29:ctrl
ID 30:ctrl
ID 31:ctrl



















U68018
MAD homolog 2
0.13
0.71
11.2
2.2
8.0
2.3
6.7
25.6


NM_016015
CGI-68 protein
0.92
1.59
2.3
2.3
3.5
3.7
3.4
22.9


AF071510
lecithin retinol
0.07
0.05
15.4
10.3
5.3
44.1
2.1
21.2



acyltransferase


AC005154
unkown
0.17
1.13
2.7
7.2
12.6
6.4
3.3
20.6


M81933
cell division
0.13
0.21
4.3
3.1
3.2
4.3
5.6
18.2



cycle 25A


AF124735
LIM HOX
0.17
0.21
2.1
4.4
5.9
5.2
7.6
17.0



gene 2


AL110125
unknown
0.30
0.08
5.0
2.7
6.8
10.2
2.8
12.0


NM_004732
potassium
0.15
0.16
7.6
4.0
3.4
2.2
2.9
11.4



voltage-gated



channel


AF030555
fatty-acid-
0.10
0.39
10.5
2.2
6.4
3.0
5.1
10.7



Coenzyme A



ligase_long-



chain 4


AF000237
1-acylglycerol-
1.80
2.37
3.4
2.5
2.4
2.1
3.7
9.9



3-phosphate O-



acyltransferase 2


AL031588
hypothetical
0.40
0.26
5.8
20.2
2.8
4.7
5.6
9.1



protein


AL080077
unknown
0.15
0.21
2.4
2.0
11.9
3.8
2.3
8.7


NM_014366
putative
0.90
2.52
2.4
4.3
2.4
2.6
3.0
8.6



nucleotide



binding



protein_estradiol-



induced


AB002359
phosphoribosyl
0.81
2.12
3.2
2.7
5.5
2.5
2.8
6.9



formylglycina



midine



synthase


U33547
MHC class II
0.14
0.16
2.5
5.3
4.5
5.0
3.1
6.6



antigen HLA-



DRB6 mRNA


AL133051
unknown
0.09
0.07
7.7
6.3
5.4
23.1
5.4
6.5


AK000576
hypothetical
0.27
0.06
7.1
9.3
5.0
6.9
2.9
6.2



protein


AF042378
spindle pole
0.36
0.39
3.3
3.0
9.5
4.5
3.4
6.2



body protein


AF093265
Homer
0.67
0.53
2.7
13.3
6.5
5.0
2.9
6.2



neuronal



immediate



early gene_3


D80000
Segregation of
1.01
1.56
3.6
2.5
4.9
3.2
6.3
6.1



mitotic



chromosomes 1


AF035309
proteasome
3.61
4.71
2.7
6.6
5.2
4.9
2.7
6.0



26S subunit



ATPase 5


M34175
adaptor-related
4.57
5.13
3.2
3.1
4.0
4.6
2.7
6.0



protein



complex 2 beta



1 subunit


AB020659
KIAA0852
0.18
0.37
4.1
7.6
5.7
4.8
2.5
5.7



protein


NM_004862
LPS-induced
2.61
3.36
3.8
4.8
4.1
4.9
3.2
5.6



TNF-alpha



factor


U00115
zinc finger
0.51
0.07
18.9
2.2
3.5
7.2
21.2
5.6



protein 51


AF088868
fibrousheathin
0.45
0.20
4.7
10.0
3.2
6.4
6.0
5.6



II


AK001890
unknown
0.42
0.55
2.4
3.5
3.6
2.3
2.2
5.6


AL137268
KIAA0759
0.49
0.34
3.8
2.3
5.0
3.5
3.3
5.4



protein


X63563
polymerase II
1.25
1.68
2.5
8.1
3.4
4.8
5.2
5.4



polypeptide B


D12676
CD36 antigen
0.35
0.39
2.9
3.4
2.6
2.2
3.5
5.3


AK000161
hypothetical
1.06
0.55
3.4
8.7
2.1
6.7
2.9
5.1



protein


AF052138
unknown
0.64
0.51
2.9
2.8
2.7
5.2
3.6
5.0


AL096803
unknown
0.36
0.03
20.1
18.3
3.7
19.3
16.1
4.9


S49953
DNA-binding
0.70
0.15
3.7
4.0
2.1
6.6
4.0
4.8



transcriptional



activator


X89399
RAS p21
0.25
0.10
8.5
14.9
4.8
18.6
4.3
4.8



protein



activator


AJ005273
antigenic
0.70
0.10
7.6
11.1
2.8
9.9
12.0
4.6



determinant of



recA protein


AK001154
hypothetical
1.70
0.96
2.4
4.4
2.9
8.9
2.4
4.5



protein


AL133605
unknown
0.26
0.15
12.4
4.2
4.4
3.3
3.3
4.1


U71092
G protein-
0.53
0.06
19.0
9.1
2.2
12.0
3.3
4.1



coupled



receptor 24


AF074723
RNA
0.67
0.54
4.0
3.2
3.1
3.4
6.0
4.0



polymerase II



transcriptional



regulation



mediator


AL137577
unknown
0.32
0.12
31.4
6.2
5.3
10.1
25.3
3.9


AF151043
hypothetical
0.48
0.35
2.6
2.2
2.0
3.3
2.2
3.8



protein


AF131831
unknown
0.67
0.81
2.1
7.0
3.5
3.2
3.9
3.7


D50405
histone
1.52
2.62
3.1
7.2
2.9
4.1
2.8
3.7



deacetylase 1


U78305
protein
1.21
0.20
4.7
13.0
3.5
5.9
4.2
3.7



phosphatase



1D


AL035562
paired box
0.24
0.01
30.2
81.9
5.6
82.3
6.2
3.7



gene 1


U67156
mitogen-
1.15
0.30
6.6
3.0
2.2
2.3
2.5
3.6



activated



protein kinase



kinase kinase 5


AL031121
unknown
0.24
0.09
5.2
3.7
2.3
6.5
9.1
3.6


U13666
G protein-
0.34
0.14
3.8
5.4
3.1
3.3
2.8
3.6



coupled



receptor 1


AB018285
KIAA0742
0.53
0.13
14.9
13.9
5.9
18.5
15.2
3.5



protein


D42053
site-1 protease
0.63
0.40
2.6
7.1
5.6
9.2
2.6
3.5


AK001135
Sec23-
0.29
0.53
5.7
4.5
3.4
2.6
11.3
3.4



interacting



protein p125


AL137461
unknown
0.25
0.02
23.8
9.0
2.7
59.2
12.5
3.3


NM_006963
zinc finger
0.10
0.08
3.2
7.6
3.7
7.9
11.2
3.2



protein 22


AL137540
unknown
0.67
0.79
3.9
2.6
5.6
4.2
3.5
3.1


AL137718
unknown
0.95
0.18
4.7
8.0
4.0
13.3
3.0
3.1


AF012086
RAN binding
1.20
0.59
4.6
4.0
2.0
4.6
3.6
3.1



protein 2-like 1


S57296
HER2/neu
0.59
0.17
7.3
12.1
2.3
20.0
22.2
3.0



receptor


NM_013329
GC-rich
0.16
0.08
6.9
14.3
9.7
3.3
7.2
3.0



sequence



DNA-binding



factor



candidate


AF038664
UDP-Gal:beta
0.15
0.03
13.4
22.2
5.4
15.8
17.6
3.0



GlcNAc beta



1_4-



galactosyltransferase


AF080579
Homo sapiens
0.34
1.03
3.3
3.0
6.7
2.1
2.9
2.9



integral



membrane



protein


AK001075
hypothetical
0.67
0.10
2.1
2.6
2.6
8.9
2.2
2.9



protein


AB011124
KIAA0552
0.46
0.04
9.6
72.0
6.0
33.9
13.6
2.9



gene product


J03068
N-
0.54
0.21
2.2
5.0
2.4
5.2
3.6
2.8



acylaminoacyl-



peptide



hydrolase


D87120
osteoblast
0.87
0.87
2.2
2.0
4.7
2.3
2.0
2.8



protein


AB006537
IL-1R
0.17
0.07
2.9
7.0
14.5
5.3
6.6
2.8



accessory



protein


L34587
transcription
2.49
1.23
2.2
16.3
5.0
15.8
5.5
2.7



elongation



factor B


D31891
SET domain_bifurcated_1
1.02
0.29
3.9
6.0
4.3
4.9
6.6
2.7


D00760
proteasome
4.97
4.94
4.1
2.6
2.0
2.8
2.7
2.7



subunit_alpha



type_2


AC004774
distal-less
0.25
0.12
2.3
6.3
3.8
5.2
5.2
2.6



homeo box 5


AL024493
unknown
1.46
0.54
4.8
13.5
2.1
11.6
6.8
2.6


AB014536
copine III
1.80
1.29
3.2
9.5
3.8
6.8
2.6
2.6


X59770
IL-1R type II
0.59
0.16
9.6
4.7
3.9
3.2
4.9
2.5


AF052183
unknown
0.65
0.76
4.0
3.7
2.3
5.0
3.0
2.5


AK000541
hypothetical
0.92
0.27
4.5
13.9
3.6
18.1
4.3
2.5



protein


U88528
cAMP
1.37
0.86
3.1
5.4
2.1
2.8
2.1
2.4



responsive



element



binding protein


M97925
defensin alpha
0.33
0.07
4.6
35.9
2.0
7.8
6.5
2.4



5_Paneth cell-



specific


NM_013393
cell division
1.38
0.94
3.1
5.8
2.1
4.2
2.6
2.3



protein FtsJ


X62744
MHC class II
0.86
0.32
4.0
4.7
2.3
2.9
6.1
2.3



DM alpha


AF251040
putative
0.64
0.30
6.7
3.4
2.9
3.9
5.7
2.2



nuclear protein


AK000227
hypothetical
1.49
0.43
3.4
7.1
2.3
3.3
9.1
2.1



protein


U88666
SFRS protein
1.78
0.37
3.4
5.9
2.6
8.4
6.1
2.0



kinase 2







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides.





Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04).





The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labeling of cDNA with the dyes Cy3 and Cy5 respectively.





The “ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.














TABLE 35










Up-regulation of Polynucleotide expression in A549 cells induced by Formula E


Peptides.
















Accession

ctrl-
ctrl-








Number
Gene
Cy3
Cy5
ID 33:ctrl
ID 34:ctrl
ID 35:ctrl
ID 36:ctrl
ID 37:ctrl
ID 38:ctrl



















AL049689
Novel human
0.25
0.05
2.7
26.5
3.3
21.7
5.4
37.9



mRNA


AK000576
hypothetical
0.27
0.06
3.0
19.1
3.9
23.0
3.1
28.3



protein


X74837
mannosidase,
0.10
0.07
5.6
10.0
10.8
12.3
12.0
19.9



alpha class



1A member 1


AK000258
hypothetical
0.27
0.07
14.0
11.1
7.9
16.1
6.2
18.9



protein


X89067
transient
0.20
0.14
3.7
2.2
2.4
2.6
8.0
18.1



receptor


AL137619
unknown
0.16
0.08
6.3
6.7
10.8
10.5
7.9
16.5


NM_003445
zinc finger
0.17
0.07
4.0
23.6
2.9
13.6
4.3
14.4



protein


X03084
complement
0.36
0.15
2.4
3.1
2.9
7.7
3.4
13.7



component 1


U27330
fucosyltransferase 5
0.39
0.08
2.4
2.5
2.6
12.1
3.5
13.0


AF070549
unknown
0.16
0.09
2.7
4.7
7.9
10.3
4.2
12.6


AB020335
sel-1-like
0.19
0.24
2.9
2.6
2.0
7.3
4.7
12.4


M26901
renin
0.09
0.12
14.9
2.2
7.3
12.0
20.8
12.0


Y07828
ring finger
0.09
0.06
9.0
26.6
8.9
16.0
3.6
11.6



protein


AK001848
hypothetical
0.21
0.07
6.2
8.2
2.7
5.2
5.5
10.9



protein


NM_016331
zinc finger
0.16
0.08
7.6
5.1
7.0
25.5
5.5
10.9



protein


U75330
neural cell
0.42
0.08
2.5
3.6
2.0
5.8
6.2
9.9



adhesion



molecule 2


AB037826
unknown
0.16
0.11
3.8
6.0
3.4
13.4
6.0
9.8


M34041
adrenergic
0.30
0.13
4.5
4.5
3.7
8.6
5.6
9.8



alpha-2B-



receptor


D38449
putative G
0.18
0.09
2.3
25.8
11.7
2.3
3.2
9.5



protein



coupled



receptor


AJ250562
transmembrane
0.13
0.10
10.0
8.4
2.2
8.1
16.3
9.1



4 superfamily



member 2


AK001807
hypothetical
0.18
0.12
4.2
5.3
4.6
3.2
4.0
8.3



protein


AL133051
unknown
0.09
0.07
5.1
13.6
6.0
9.1
2.2
8.2


U43843
Neuro-d4
0.61
0.10
2.0
6.4
2.3
16.6
2.2
8.1



homolog


NM_013227
aggrecan 1
0.28
0.15
7.5
3.1
2.5
6.9
8.5
7.8


AF226728
somatostatin
0.23
0.17
7.0
3.6
3.1
5.5
3.5
7.7



receptor-



interacting



protein


AK001024
guanine
0.16
0.11
3.9
12.3
2.7
7.4
3.3
7.0



nucleotide-



binding



protein


AC002302
unknown
0.13
0.14
16.1
5.8
5.8
2.6
9.6
6.2


AB007958
unknown
0.17
0.27
2.0
2.3
11.3
3.3
3.0
6.1


AF059293
cytokine
0.19
0.22
3.6
2.5
10.2
3.8
2.7
5.9



receptor-like



factor 1


V01512
v-fos
0.27
0.21
6.7
3.7
13.7
9.3
3.7
5.4


U82762
sialyltransferase 8
0.23
0.15
3.2
6.5
2.7
9.2
5.7
5.4


U44059
thyrotrophic
0.05
0.13
22.9
7.1
12.5
7.4
9.7
5.4



embryonic



factor


X05323
antigen
0.39
0.13
4.3
2.5
2.2
7.4
2.8
5.1



identified by



monoclonal



antibody


U72671
ICAM 5,
0.25
0.14
5.3
2.7
3.7
10.0
3.2
4.8


AL133626
hypothetical
0.26
0.25
2.2
4.2
2.9
3.0
2.6
4.7



protein


X96401
MAX
0.31
0.29
6.9
2.3
4.9
3.1
2.9
4.6



binding



protein


AL117533
unknown
0.05
0.26
8.2
2.7
11.1
2.5
11.9
4.5


AK001550
hypothetical
0.10
0.30
8.0
2.0
4.9
2.1
7.8
4.5



protein


AB032436

Homo

0.14
0.21
5.1
2.2
9.1
4.5
6.4
4.4




sapiens BNPI




mRNA


AL035447
hypothetical
0.28
0.23
4.3
3.7
8.7
5.2
3.7
4.2



protein


U09414
zinc finger
0.28
0.25
4.0
2.2
4.7
3.3
7.2
4.2



protein


AK001256
unknown
0.09
0.08
5.3
6.5
31.1
12.7
6.4
4.1


L14813
carboxyl
0.64
0.21
2.7
6.2
3.1
2.1
3.4
3.9



ester lipase-



like


AF038181
unknowan
0.06
0.18
34.1
6.4
4.5
8.7
11.3
3.9


NM_001486
glucokinase
0.21
0.08
3.0
2.2
6.5
12.4
5.7
3.9


AB033000
hypothetical
0.24
0.22
3.4
3.3
7.1
5.5
4.5
3.8



protein


AL117567
DKFZP566O
0.44
0.22
2.2
2.7
3.9
4.0
4.5
3.7



084 protein


NM_012126
carbohydrate
0.31
0.20
5.5
5.4
3.8
5.5
2.6
3.5



sulfotransferase 5


AL031687
unknown
0.16
0.27
5.9
2.6
3.4
2.3
4.9
3.5


X04506
apolipoprotein B
0.29
0.32
5.4
4.4
6.9
5.5
2.1
3.5


NM_006641
CCR9
0.35
0.11
3.3
3.3
2.2
16.5
2.3
3.5


Y00970
acrosin
0.12
0.14
8.2
8.8
3.1
6.2
17.5
3.4


X67098
rTS beta
0.19
0.26
2.4
3.1
7.8
3.5
4.4
3.3



protein


U51990
pre-mRNA
0.56
0.19
2.2
3.0
2.8
13.7
2.9
3.0



splicing



factor


AF030555
fatty-acid-
0.10
0.39
3.5
6.9
13.3
4.4
7.5
2.9



Coenzyme A


AL009183
TNFR
0.46
0.19
6.0
4.1
2.8
8.6
2.6
2.8



superfamily,



member 9


AF045941
sciellin
0.16
0.21
11.6
2.4
2.8
2.2
4.1
2.8


AF072756
A kinase
0.33
0.07
2.5
5.3
3.9
32.7
2.3
2.7



anchor



protein 4


X78678
ketohexokinase
0.10
0.20
18.0
3.5
4.1
2.5
14.6
2.6


AL031734
unknown
0.03
0.39
43.7
2.3
41.7
4.0
10.8
2.5


D87717
KIAA0013
0.35
0.42
4.2
2.3
3.6
2.6
2.9
2.5



gene product


U01824
solute carrier
0.42
0.29
4.8
2.3
4.2
7.1
4.2
2.4



family 1


AF055899
solute carrier
0.14
0.31
9.5
12.3
7.4
4.7
6.6
2.3



family 27


U22526
lanosterol
0.09
0.45
4.1
3.4
10.4
2.2
17.9
2.3



synthase


AB032963
unknown
0.19
0.34
6.3
6.1
2.9
2.1
5.7
2.2


NM_015974
lambda-
0.17
0.25
11.4
2.8
5.9
2.4
5.8
2.2



crystallin


X82200
stimulated
0.23
0.15
8.2
3.4
3.0
2.8
11.3
2.2



trans-acting



factor


AL137522
unknown
0.12
0.26
12.1
3.7
12.6
6.9
4.3
2.2


Z99916
crystallin,
0.28
0.65
2.5
2.1
3.6
2.2
2.6
2.1



beta B3


AF233442
ubiquitin
0.41
0.31
2.6
3.6
3.6
4.5
3.4
2.1



specific



protease 21


AK001927
hypothetical
0.24
0.52
7.6
5.6
5.0
2.5
4.1
2.0



protein







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides.





Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04).





The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labeling of cDNA with the dyes Cy3 and Cy5 respectively.





The “ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.














TABLE 36










Up-regulation of Polynucleotide expression in A549 cells induced by Formula F


Peptides.















Accession

ctrl-
ctrl-
Ratio
Ratio
Ratio
Ratio
Ratio


Number
Gene
Cy3
Cy5
ID 40:ctrl
ID 42:ctrl
ID 43:ctrl
ID 44:ctrl
ID 45:ctrl


















AF025840
polymerase
0.34
0.96
3.4
2.0
2.0
2.1
4.3



epsilon 2


AF132495
CGI-133
0.83
0.67
3.0
2.2
2.6
2.8
5.1



protein


AL137682
hypothetical
0.73
0.40
2.0
5.3
4.8
2.9
8.2



protein


U70426
regulator of
0.23
0.25
3.1
3.0
5.3
3.1
12.2



G-protein



signaling 16


AK001135
Sec23-
0.29
0.53
3.2
2.6
3.3
14.4
5.2



interacting



protein



p125


AB023155
KIAA0938
0.47
0.21
2.7
4.8
8.1
4.2
10.4



protein


AB033080
cell cycle
0.31
0.31
4.4
2.2
5.9
4.3
6.9



progression



8 protein


AF061836
Ras
0.29
0.31
3.2
2.5
11.1
18.8
6.8



association



domain



family 1


AK000298
hypothetical
0.48
0.27
3.3
2.2
7.1
5.6
7.7



protein


L75847
zinc finger
0.35
0.52
3.2
3.0
4.0
3.0
3.9



protein


X97267
protein
0.19
0.24
4.1
9.3
2.4
4.2
8.3



tyrosine



phosphatase


Z11933
POU
0.09
0.23
8.7
2.5
3.6
4.3
8.2



domain



class 3 TF 2


AB037744
unknown
0.37
0.57
2.6
2.9
2.7
3.0
3.1


U90908
unknown
0.12
0.16
11.8
7.7
3.4
7.8
11.2


AL050139
unknown
0.29
0.60
5.2
2.4
3.3
3.0
2.8


AB014615
fibroblast
0.19
0.07
5.4
3.5
8.5
3.2
22.7



growth



factor 8


M28825
CD1A
0.51
0.36
4.1
2.6
2.0
4.6
4.4



antigen


U27330
fucosyltransferase 5
0.39
0.08
3.3
2.1
24.5
8.2
19.3


NM_00696
zinc finger
0.10
0.08
10.4
12.6
12.3
29.2
20.5



protein


AF093670
peroxisomal
0.44
0.53
4.0
2.6
2.6
4.3
2.9



biogenesis



factor


AK000191
hypothetical
0.50
0.182
2.3
3.6
4.4
2.2
8.2



protein


AB022847
unknown
0.39
0.24
2.1
6.9
4.5
2.8
6.2


AK000358
microfibrillar-
0.28
0.28
5.7
2.0
3.5
5.2
5.2



associated



protein 3


X74837
mannosidase_alpha
0.10
0.07
13.1
18.4
23.6
16.3
20.8



class 1A


AF053712
TNF
0.17
0.08
11.3
9.3
13.4
10.6
16.6



superfamily_member



11


AL133114
DKFZP586
0.11
0.32
8.5
3.4
4.9
5.3
4.3



P2421



protein


AF049703
E74-like
0.22
0.24
5.1
6.0
3.3
2.7
5.4



factor 5


AL137471
hypothetical
0.29
0.05
4.0
15.0
10.1
2.7
25.3



protein


AL035397
unknown
0.33
0.14
2.3
2.8
10.6
4.6
9.3


AL035447
hypothetical
0.28
0.23
3.8
6.8
2.7
3.0
5.7



protein


X55740
CD73
0.41
0.61
2.1
3.3
2.9
3.2
2.1


NM_004909
taxol
0.20
0.22
3.9
2.9
6.5
3.2
5.6



resistance



associated



gene 3


AF233442
ubiquitin
0.41
0.31
2.9
4.7
2.7
3.5
3.9



specific



protease


U92980
unknown
0.83
0.38
4.2
4.1
4.8
2.3
3.1


AF105424
myosin
0.30
0.22
2.8
3.3
4.4
2.3
5.3



heavy



polypeptide-



like


M26665
histatin 3
0.29
0.26
7.9
3.5
4.6
3.5
4.5


AF083898
neuro-
0.20
0.34
18.7
3.8
2.2
3.6
3.5



oncological



ventral



antigen 2


AJ009771
ariadne_Drosophila_homolog
0.33
0.06
2.3
17.6
15.9
2.5
20.3



of


AL022393
hypothetical
0.05
0.33
32.9
2.4
3.0
69.4
3.4



protein P1


AF039400
chloride
0.11
0.19
8.4
2.9
5.1
18.1
5.9



channel_calcium



activated_family



member 1


AJ012008
dimethylarginine
0.42
0.43
5.1
3.3
3.2
6.2
2.6



dimethylaminohydrolase 2


AK000542
hypothetical
0.61
0.24
2.1
4.5
5.0
3.7
4.4



protein


AL133654
unknown
0.27
0.40
2.8
2.1
2.5
2.5
2.6


AL137513
unknown
0.43
0.43
6.4
3.2
3.8
2.3
2.3


U05227
GTP-
0.38
0.36
5.0
3.1
3.1
2.2
2.8



binding



protein


D38449
putative G
0.18
0.09
5.8
6.7
6.7
9.1
10.4



protein



coupled



receptor


U80770
unknown
0.31
0.14
3.9
3.8
6.6
3.1
6.8


X61177
IL-5R alpha
0.40
0.27
2.6
4.4
9.8
8.1
3.6


U35246
vacuolar
0.15
0.42
5.8
2.8
2.6
4.5
2.2



protein



sorting 45A


AB017016
brain-
0.27
0.29
6.0
2.6
3.4
3.1
3.1



specific



protein p25



alpha


X82153
cathepsin K
0.45
0.20
4.2
5.2
4.8
4.4
4.6


AC005162
probable
0.12
0.28
11.9
3.4
6.8
18.7
3.2



carboxypeptidase



precursor


AL137502
unknown
0.22
0.16
3.9
4.9
7.3
3.9
5.3


U66669
3-
0.30
0.40
10.3
3.5
5.2
2.3
2.1



hydroxyisobutyryl-



Coenzyme



A hydrolase


AK000102
unknown
0.39
0.30
2.8
5.3
5.2
4.1
2.8


AF034970
docking
0.28
0.05
3.3
8.5
15.7
4.0
17.3



protein 2


AK000534
hypothetical
0.13
0.29
6.8
2.3
4.0
20.6
2.9



protein


J04599
biglycan
0.39
0.30
4.0
3.7
4.0
4.8
2.8


AL133612
unknown
0.62
0.33
2.7
3.4
5.2
3.0
2.5


D10495
protein
0.18
0.10
12.0
20.7
8.7
6.8
8.1



kinase C



delta


X58467
cytochrome
0.07
0.24
15.4
4.7
7.9
34.4
3.4



P450


AF131806
unknown
0.31
0.25
2.6
3.4
5.7
7.0
3.2


AK000351
hypothetical
0.34
0.13
4.0
6.9
5.5
2.8
6.3



protein


AF075050
hypothetical
0.55
0.09
2.7
17.8
5.1
2.2
8.3



protein


AK000566
hypothetical
0.15
0.35
6.7
2.2
6.8
6.4
2.1



protein



unknown


U43328
cartilage
0.44
0.19
2.5
6.2
6.9
7.8
3.8



linking



protein 1


AF045941
sciellin
0.16
0.21
6.8
7.5
4.8
6.9
3.4


U27655
regulator of
0.24
0.29
5.5
4.9
2.9
4.9
2.4



G-protein



signaling 3


AK000058
hypothetical
0.25
0.15
5.0
9.7
16.4
2.7
4.5



protein


AL035364
hypothetical
0.32
0.26
4.4
4.2
7.3
2.8
2.6



protein


AK001864
unknown
0.40
0.25
3.7
3.7
4.6
3.2
2.6


AB015349
unknown
0.14
0.24
10.5
2.8
3.7
8.0
2.7


V00522
MHC class
0.62
0.22
4.8
3.9
4.7
2.5
3.0



II DR beta 3


U75330
neural cell
0.42
0.08
2.1
9.6
13.2
3.3
7.8



adhesion



molecule 2


NM_007199
IL-1R-
0.15
0.25
8.7
7.8
8.6
16.1
2.5



associated



kinase M


D30742
calcium/cal
0.28
0.09
6.2
28.7
7.4
2.4
6.8



modulin-



dependent



protein



kinase IV


X05978
cystatin A
0.63
0.17
2.7
4.8
9.4
2.2
3.6


AF240467
TLR-7
0.11
0.10
13.8
13.3
4.7
7.7
4.9







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides.





Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Human Operon arrays (PRHU04).





The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labeling of cDNA with the dyes Cy3 and CyS respectively.





The “Ratio ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.














TABLE 37










Up-regulation of Polynucleotide expression in A549 cells induced by Formula G


and additional Peptides.

















Accession
ctrl-
ctrl-










Number
Cy3
Cy5
ID 53:ctrl
ID 54:ctrl
ID 47:ctrl
ID 48:ctrl
ID 49:ctrl
ID 50:ctrl
ID 51:ctrl
ID 52:ctrl




















U00115
0.51
0.07
27.4
7.3
2.4
3.1
4.8
8.3
3.5
20.0


M91036
0.22
0.02
39.1
32.5
5.2
2.2
37.0
6.0
16.2
18.0


AK000070
0.36
0.18
3.8
7.6
2.6
15.1
12.2
9.9
17.2
15.3


AF055899
0.14
0.31
6.7
3.7
9.7
10.0
2.2
16.7
5.4
14.8


AK001490
0.05
0.02
14.1
35.8
3.2
28.6
25.0
20.2
56.5
14.1


X97674
0.28
0.28
3.2
3.7
4.0
10.7
3.3
3.1
4.0
13.2


AB022847
0.39
0.24
4.1
4.4
4.5
2.7
3.7
10.4
5.0
11.3


AJ275986
0.26
0.35
5.8
2.3
5.7
2.2
2.5
9.7
4.3
11.1


D10495
0.18
0.10
8.0
3.4
4.6
2.0
6.9
2.5
12.7
10.3


L36642
0.26
0.06
5.8
14.2
2.6
4.1
8.9
3.4
6.5
6.6


M31166
0.31
0.12
4.8
3.8
12.0
3.6
9.8
2.4
8.8
6.4


AF176012
0.45
0.26
3.1
2.9
2.8
2.6
2.3
6.9
3.0
5.8


AF072756
0.33
0.07
9.9
9.3
4.4
4.3
3.2
4.9
11.9
5.4


NM_014439
0.47
0.07
12.0
7.1
3.3
3.3
4.7
5.9
5.0
5.4


AJ271351
0.46
0.12
3.4
3.5
2.3
4.7
2.3
2.7
6.9
5.2


AK000576
0.27
0.06
7.4
15.7
2.9
4.7
9.0
2.4
8.2
5.1


AJ272265
0.21
0.09
6.2
7.9
2.3
3.7
10.3
4.5
4.6
4.7


AL122038
0.46
0.06
6.7
4.5
2.6
4.3
16.4
6.5
26.6
4.6


AK000307
0.23
0.09
3.7
4.0
4.3
3.2
5.3
2.9
13.1
4.4


AB029001
0.52
0.21
14.4
4.3
4.6
4.4
4.8
21.9
3.2
4.2


U62437
0.38
0.13
12.6
6.5
4.2
6.7
2.2
3.7
4.8
3.9


AF064854
0.15
0.16
2.6
2.9
6.2
8.9
14.4
5.0
9.1
3.9


AL031588
0.40
0.26
8.3
5.2
2.8
3.3
5.3
9.0
5.6
3.4


X89399
0.25
0.10
15.8
12.8
7.4
4.2
16.7
6.9
12.7
3.3


D45399
0.21
0.18
3.0
4.7
3.3
4.4
8.7
5.3
5.1
3.3


AB037716
0.36
0.40
5.1
7.5
2.6
2.1
3.5
3.1
2.4
2.8


X79981
0.34
0.10
4.7
7.2
3.2
4.6
6.5
5.1
5.8
2.7


AF034208
0.45
0.24
2.7
10.9
2.1
3.7
2.3
5.9
2.2
2.5


AL133355
0.22
0.23
2.3
3.4
7.3
2.7
3.3
4.3
2.8
2.5


NM_016281
0.40
0.19
6.6
10.6
2.1
2.8
5.0
11.2
10.6
2.5


AF023614
0.11
0.42
2.2
2.2
6.0
7.5
5.0
2.7
2.0
2.4


AF056717
0.43
0.62
4.3
3.2
5.1
4.0
4.6
9.7
3.1
2.2


AB029039
0.79
0.49
2.7
3.3
3.7
2.0
2.3
2.4
4.8
2.2


J03634
0.40
0.12
3.7
2.3
2.3
4.0
10.5
4.1
9.1
2.2


U80764
0.31
0.18
2.3
7.4
4.2
2.3
5.1
3.3
8.8
2.1


AB032963
0.19
0.34
4.0
7.3
5.0
3.0
2.9
6.7
3.8
2.1


X82835
0.25
0.38
2.0
2.7
2.9
7.7
3.3
3.1
3.5
2.0







The peptides at a concentration of 50 μg/ml were shown to increase the expression of many polynucleotides.





Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labelled cDNA probes and hybridised to Human Operon arrays (PRHU04).





The intensity of polynucleotides in control, unstimulated cells are shown in the second and third columns for labelling of cDNA with the dyes Cy3 and Cy5 respectively.





The “Ratio ID#: Control” columns refer to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.





Accession numbers and gene designations are U00115, zinc finger protein; M91036, hemoglobin gamma G; K000070, hypothetical protein; AF055899, solute carrier family 27; AK001490, hypothetical protein; X97674, nuclear receptor coactivator 2; AB022847, unknown; AJ275986, transcription factor; D10495, protein kinase C, delta; L36642, EphA7; M31166, pentaxin-related gene; AF176012,



# unknown; AF072756, A kinase anchor protein 4; NM_014439, IL-1 Superfamily z; AJ271351, putative transcriptional regulator; AK000576, hypothetical protein; AJ272265, secreted phosphoprotein 2; AL122038, hypothetical protein; AK000307, hypothetical protein; AB029001, KIAA1078 protein; U62437, cholinergic receptor; AF064854, unknown; AL031588, hypothetical protein; X89399, RAS p21 protein activator; D45399,


# phosphodiesterase; AB037716, hypothetical protein; X79981, cadherin 5; AF034208, RIG-like 7-1; AL133355, chromosome 21 open reading frame 53; NM_016281, STE20-like kinase; AF023614, transmembrane activator and CAML interactor; AF056717, ash2-like; AB029039, KIAA1116 protein; J03634, inhibin, beta A; U80764, unknown; AB032963, unknown; X82835, sodium channel, voltage-gated, type IX.






EXAMPLE 5
Induction of Chemokines in Cell Lines, Whole Human Blood, and in Mice by Peptides

The murine macrophage cell line RAW 264.7, THP-1 cells (human monocytes), a human epithelial cell line (A549), human bronchial epithelial cells (16HBEo14), and whole human blood were used. HBE cells were grown in MEM with Earle's. THP-1 cells were grown and maintained in RPMI 1640 medium. The RAW and A549 cell lines were maintained in DMEM supplemented with 10% fetal calf serum. The cells were seeded in 24 well plates at a density of 106 cells per well in DMEM (see above) and A549 cells were seeded in 24 well plates at a density of 105 cells per well in DMEM (see above) and both were incubated at 37° C. in 5% CO2 overnight. DMEM was aspirated from cells grown overnight and replaced with fresh medium. After incubation of the cells with peptide, the release of chemokines into the culture supernatant was determined by ELISA (R&D Systems, Minneapolis, Minn.).


Animal studies were approved by the UBC Animal Care Committee.(UBC ACC # A01-0008). BALB/c mice were purchased from Charles River Laboratories and housed in standard animal facilities. Age, sex and weight matched adult mice were anaesthetized with an intraperitoneal injection of Avertin (4.4 mM 2-2-2-tribromoethanol, 2.5% 2-methyl-2-butanol, in distilled water), using 200 ill per 10 g body weight. The instillation was performed using a non-surgical, intratracheal instillation method adapted from Ho and Furst 1973. Briefly, the anaesthetized mouse was placed with its upper teeth hooked over a wire at the top of a support frame with its jaw held open and a spring pushing the thorax forward to position the pharynx, larynx and trachea in a vertical straight line. The airway was illuminated externally and an intubation catheter was inserted into the clearly illuminated tracheal lumen. Twenty-μl of peptide suspension or sterile water was placed in a well at the proximal end of the catheter and gently instilled into the trachea with 200 μl of air. The animals were maintained in an upright position for 2 minutes after instillation to allow the fluid to drain into the respiratory tree. After 4 hours the mice were euthanaised by intraperitoneal injection of 300 mg/kg of pentobarbital. The trachea was exposed; an intravenous catheter was passed into the proximal trachea and tied in place with suture thread. Lavage was performed by introducing 0.75 ml sterile PBS into the lungs via the tracheal cannula and then after a few seconds, withdrawing the fluid. This was repeated 3 times with the same sample of PBS. The lavage fluid was placed in a tube on ice and the total recovery volume per mouse was approximately 0.5 ml. The bronchoalveolar.lavage (BAL) fluid was centrifuged at 1200 rpm for 10 min, the clear supernatant removed and tested for TNF-α and MCP-1 by ELISA.


The up-regulation of chemokines by cationic peptides was confirmed in several different systems. The murine MCP-1, a homologue of the human MCP-1, is a member of the β(C-C) chemokine family. MCP-1 has been demonstrated to recruit monocytes, NK cells and some T lymphocytes. When RAW 264.7 macrophage cells and whole human blood from 3 donors were stimulated with increasing concentrations of peptide, SEQ ID NO: 1, they produced significant levels of MCP-1 in their supernatant, as judged by ELISA (Table 36). RAW 264.7 cells stimulated with peptide concentrations ranging from 20-50 μg/ml for 24 hr produced significant levels of MCP-1(200-400 pg/ml above background). When the cells (24 h) and whole blood (4 h) were stimulated with 100 μg/ml of SEQ ID NO: 1, high levels of MCP-1 were produced.


The effect of cationic peptides on chemokine induction was also examined in a completely different cell system, A549 human epithelial cells. Interestingly, although these cells produce MCP-1 in response to LPS, and this response could be antagonized by peptide; there was no production of MCP-1 by A549 cells in direct response to peptide, SEQ ID NO: 1. Peptide SEQ ID NO: 1 at high concentrations, did however induce production of IL-8, a neutrophil specific chemokine (Table 37). Thus, SEQ ID NO: 1 can induce a different spectrum of responses from different cell types and at different concentrations. A number of peptides from each of the formula groups were tested for their ability to induce IL-8 in A549 cells (Table 38). Many of these peptides at a low concentration, 10 μg/ml induced IL-8 above background levels. At high concentrations (100 μg/ml) SEQ ID NO: 13 was also found to induce IL-8 in whole human blood (Table 39). Peptide SEQ ID NO: 2 also significantly induced IL-8 in HBE cells (Table 40) and undifferentiated THP-1 cells (Table 41).


BALB/c mice were given SEQ ID NO: 1 or endotoxin-free water by intratracheal instillation and the levels of MCP-1 and TNF-α examined in the bronchioalveolar lavage fluid after 3-4 hr. It was found that the mice treated with 50 μg/ml peptide, SEQ ID NO: 1 produced significantly increased levels of MCP-1 over mice given water or anesthetic alone (Table 42). This was not a pro-inflammatory response to peptide, SEQ ID NO: 1 since peptide did not significantly induce more TNF-α than mice given water or anesthetic alone. peptide, SEQ ID NO: 1 was also found not to significantly induce TNF-α production by RAW 264.7 cells and bone marrow-derived macrophages treated with peptide, SEQ ID NO: 1 (up to 100 μg/ml) (Table 43). Thus, peptide, SEQ ID NO: 1 selectively induces the production of chemokines without inducing the production of inflammatory mediators such as TNF-α. This illustrates the dual role of peptide, SEQ ID NO: 1 as a factor that can block bacterial product-induced inflammation while helping to recruit phagocytes that can clear infections.

TABLE 38Induction of MCP-1 in RAW 264.7 cells and whole human blood.Monocyte chemoattractantPeptide, SEQ ID NO: 1protein (MCP)-1 (pg/ml)*(μg/ml)RAW cellsWhole blood0135.3 ± 16.3112.7 ± 43.3 10165.7 ± 18.2239.3 ± 113.350  367 ± 11.5371 ± 105100  571 ± 17.4  596 ± 248.1
RAW 264.7 mouse macrophage cells or whole human blood were stimulated with increasing concentrations of SEQ ID NO: 1 for 4 hr. The human blood samples were centrifuged and the serum was removed and tested for MCP-1 by ELISA along with the supernatants from the RAW 264.7 cells. The RAW cell data presented is the mean of three or more experiments ± standard error and the human blood data represents the mean ± standard error from three separate donors.









TABLE 39










Induction of IL-8 in A549 cells and whole human blood.









Peptide, SEQ ID NO: 1
IL-8 (pg/ml)










(μg/ml)
A549 cells
Whole blood












0
  172 ± 29.1
660.7 ± 126.6


1
206.7 ± 46.1


10
283.3 ± 28.4
945.3 ± 279.9


20
  392 ± 31.7


50
542.3 ± 66.2
1160.3 ± 192.4 


100
1175.3 ± 188.3







A549 cells or whole human blood were stimulated with increasing concentrations of peptide for 24 and 4 hr respectively. The human blood samples were centrifuged and the serum was removed and tested for IL-8 by ELISA along with the supernatants from the A549 cells. The A549 cell data presented is the mean of three or more experiments ± standard error and the human blood data represents the mean ± standard error from three separate donors.














TABLE 40










Induction of IL-8 in A549 cells by Cationic peptides.










Peptide (10 ug/ml)
IL-8 (ng/ml)














No peptide
0.164



LPS, no peptide
0.26



SEQ ID NO: 1
0.278



SEQ ID NO: 6
0.181



SEQ ID NO: 7
0.161



SEQ ID NO: 9
0.21



SEQ ID NO: 10
0.297



SEQ ID NO: 13
0.293



SEQ ID NO: 14
0.148



SEQ ID NO: 16
0.236



SEQ ID NO: 17
0.15



SEQ ID NO: 19
0.161



SEQ ID NO: 20
0.151



SEQ ID NO: 21
0.275



SEQ ID NO: 22
0.314



SEQ ID NO: 23
0.284



SEQ ID NO: 24
0.139



SEQ ID NO: 26
0.201



SEQ ID NO: 27
0.346



SEQ ID NO: 28
0.192



SEQ ID NO: 29
0.188



SEQ ID NO: 30
0.284



SEQ ID NO: 31
0.168



SEQ ID NO: 33
0.328



SEQ ID NO: 34
0.315



SEQ ID NO: 35
0.301



SEQ ID NO: 36
0.166



SEQ ID NO: 37
0.269



SEQ ID NO: 38
0.171



SEQ ID NO: 40
0.478



SEQ ID NO: 41
0.371



SEQ ID NO: 42
0.422



SEQ ID NO: 43
0.552



SEQ ID NO: 44
0.265



SEQ ID NO: 45
0.266



SEQ ID NO: 47
0.383



SEQ ID NO: 48
0.262



SEQ ID NO: 49
0.301



SEQ ID NO: 50
0.141



SEQ ID NO: 51
0.255



SEQ ID NO: 52
0.207



SEQ ID NO: 53
0.377



SEQ ID NO: 54
0.133









A549 human epithelial cells were stimulated with 10 μg of peptide for 24 hr. The supernatant was removed and tested for IL-8 by ELISA.














TABLE 41










Induction by Peptide of IL-8 in human blood.










SEQ ID NO: 3 (μg/ml)
IL-8 (pg/ml)














0
85



10
70



100
323









Whole human blood was stimulated with increasing concentrations of peptide for 4 hr. The human blood samples were centrifuged and the serum was removed and tested for IL-8 by ELISA. The data shown is the average 2 donors.














TABLE 42










Induction of IL-8 in HBE cells.










SEQ ID NO: 2




(μg/ml)
IL-8 (pg/ml)














0
552 ± 90



0.1
 670 ± 155



1
 712 ± 205



10
941 ± 15



50
1490 ± 715









Increasing concentrations of the peptide were incubated with HBE cells for 8 h, the supernantant removed and tested for IL-8. The data is presented as the mean of three or more experiments ± standard error.














TABLE 43










Induction of IL-8 in undifferentiated THP-1 cells.










SEQ ID NO: 3




(μg/ml)
IL-8 (pg/ml)














0
10.6



10
17.2



50
123.7









The human monocyte THP-1 cells were incubated with indicated concentrations of peptide for 8 hr. The supernatant was removed and tested for IL-8 by ELISA.














TABLE 44










Induction of MCP-1 by Peptide, SEQ ID NO: 1 in mouse airway.











Condition
MCP-1 (pg/ml)
TNF-α (pg/ml)







Water
16.5 ± 5  
664 ± 107



peptide
111 ± 30 
734 ± 210



Avertin
6.5 ± 0.5
393 ± 129









BALB/c mice were anaesthetised with avertin and given intratracheal instillation of peptide or water or no instillation (no treatment). The mice were monitored for 4 hours, anaesthetised and the BAL fluid was isolated and analyzed for MCP-1 and TNF-α concentrations by ELISA. The data shown is the mean of 4 or 5 mice for each condition ± standard error.














TABLE 45










Lack of Significant TNF-α induction by the Cationic Peptides.










Peptide Treatment
TNF-α (pg/ml)







Media background
56 ± 8



LPS treatment, No peptide
15207 ± 186 



SEQ ID NO: 1
274 ± 15



SEQ ID NO: 5
223 ± 45



SEQ ID NO: 6
297 ± 32



SEQ ID NO: 7
270 ± 42



SEQ ID NO: 8
166 ± 23



SEQ ID NO: 9
171 ± 33



SEQ ID NO: 10
288 ± 30



SEQ ID NO: 12
299 ± 65



SEQ ID NO: 13
216 ± 42



SEQ ID NO: 14
226 ± 41



SEQ ID NO: 15
346 ± 41



SEQ ID NO: 16
341 ± 68



SEQ ID NO: 17
249 ± 49



SEQ ID NO: 19
397 ± 86



SEQ ID NO: 20
285 ± 56



SEQ ID NO: 21
263 ± 8 



SEQ ID NO: 22
195 ± 42



SEQ ID NO: 23
254 ± 58



SEQ ID NO: 24
231 ± 32



SEQ ID NO: 26
281 ± 34



SEQ ID NO: 27
203 ± 42



SEQ ID NO: 28
192 ± 26



SEQ ID NO: 29
242 ± 40



SEQ ID NO: 31
307 ± 71



SEQ ID NO: 33
196 ± 42



SEQ ID NO: 34
204 ± 51



SEQ ID NO: 35
274 ± 76



SEQ ID NO: 37
323 ± 41



SEQ ID NO: 38
199 ± 38



SEQ ID NO: 43
 947 ± 197



SEQ ID NO: 44
 441 ± 145



SEQ ID NO: 45
398 ± 90



SEQ ID NO: 48
253 ± 33



SEQ ID NO: 49
324 ± 38



SEQ ID NO: 50
 311 ± 144



SEQ ID NO: 53
263 ± 40



SEQ ID NO: 54
346 ± 86









RAW 264.7 macrophage cells were incubated with indicated peptides (40 μg/ml) for 6 hours. The supernatant was collected and tested for levels of TNF-α by ELISA. The data is presented as the mean of three or more experiments + standard error.







EXAMPLE 6
Cationic Peptides Increase Surface Expression of Chemokine Receptors

To analyze cell surface expression of IL-8RB, CXCR-4, CCR2, and LFA-1, RAW macrophage cells were stained with 10 μg/ml of the appropriate primary antibody (Santa Cruz Biotechnology) followed by FITC-conjugated goat anti-rabbit IgG [IL-8RB and CXCR-4 (Jackson ImmunoResearch Laboratories, West Grove, Pa.)] or FITC-conjugated donkey anti-goat IgG (Santa Cruz). The cells were analyzed using a FACscan, counting 10,000 events and gating on forward and side scatter to exclude cell debris.


The polynucleotide array data suggested that some peptides up-regulate the expression of the chemokine receptors IL-8RB, CXCR-4 and CCR2 by 10, 4 and 1.4 fold above unstimulated cells respectively. To confirm the polynucleotide array data, the surface expression was examined by flow cytometry of these receptors on RAW cells stimulated with peptide for 4 hr. When 50 μg/ml of peptide was incubated with RAW cells for 4 hr, IL-8RB was upregulated an average of 2.4-fold above unstimulated cells, CXCR-4 was up-regulated an average of 1.6-fold above unstimulated cells and CCR2 was up-regulated 1.8-fold above unstimulated cells (Table 46). As a control CEMA was demonstrated to cause similar up-regulation. SEQ ID NO: 3 was the only peptide to show significant up-regulation of LFA-1 (3.8 fold higher than control cells).

TABLE 46Increased surface expression of CXCR-4, IL-8RB andCCR2 in response to peptides.ConcentrationFold Increase in Protein ExpressionPeptide(μg/ml)IL-8RBCXCR-4CCR2SEQ ID NO: 1101.01.01.0SEQ ID NO: 150 1.3 ± 0.051.3 ± 0.031.3 ± 0.03SEQ ID NO: 11002.4 ± 0.61.6 ± 0.231.8 ± 0.15SEQ ID NO: 31002.0 ± 0.6Not Done4.5CEMA501.6 ± 0.11.5 ± 0.2 1.5 ± 0.15CEMA1003.6 ± 0.8Not Done4.7 ± 1.1 
RAW macrophage cells were stimulated with peptide for 4 hr. The cells were washed and stained with the appropriate primary and FITC-labeled secondary antibodies. The data shown represents the average (fold change of RAW cells stimulated with peptide from media) ± standard error.


EXAMPLE 7
Phosphorylation of Map Kinases by Cationic Peptides

The cells were seeded at 2.5×105−5×105 cells/ml and left overnight. They were washed once in media, serum starved in the morning (serum free media—4 hrs). The media was removed and replaced with PBS, then sat at 37° C. for 15 minutes and then brought to room temp for 15 minutes. Peptide was added (concentrations 0.1 μg/ml−50 μg/ml) or H2O and incubated 10 min. The PBS was very quickly removed and replaced with ice-cold radioimmunoprecipitation (RIPA) buffer with inhibitors (NaF, B-glycerophosphate, MOL, Vanadate, PMSF, Leupeptin Aprotinin). The plates were shaken on ice for 10-15 min or until the cells were lysed and the lysates collected. The procedure for THP-1 cells was slightly different; more cells (2×106) were used. They were serum starved overnight, and to stop the reaction 1 ml of ice-cold PBS was added then they sat on ice 5-10 min, were spun down then resuspended in RIPA. Protein concentrations were determined using a protein assay (Pierce, Rockford, Ill.). Cell lysates (20 μg of protein) were separated by SDS-PAGE and transferred to nitrocellulose filters. The filters were blocked for 1 h with 10 mM Tris-HCI, pH 7.5, 150 mM NaCl (TBS)/5% skim milk powder and then incubated overnight in the cold with primary antibody in TBS/0.05% Tween 20. After washing for 30 min with TBS/0.05% Tween 20, the filters were incubated for 1 h at room temperature with 1 μg/ml secondary antibody in TBS. The filters were washed for 30 min with TBS/0.05% Tween 20 and then incubated 1 h at room temperature with horseradish peroxidase-conjugated sheep anti-mouse IgG (1:10,000 in TBS/0.05% Tween 20). After washing the filters for 30 min with TBS/0.1% Tween 20, immunoreactive bands were visualized by enhanced chemiluminescence (ECL) detection. For experiments with peripheral blood mononuclear cells: The peripheral blood (50-100 ml) was collected from all subjects. Mononuclear cells were isolated from the peripheral blood by density gradient centrifugation on Ficoll-Hypaque. Interphase cells (mononuclear cells) were recovered, washed and then resuspended in recommended primary medium for cell culture (RPMI-1640) with 10% fetal calf serum (FCS) and 1% L-glutamine. Cells were added to 6 well culture plates at 4×106 cells/well and were allowed to adhere at 37° C. in 5% CO2 atmosphere for 1 hour. The supernatant medium and non-adherent cells were washed off and the appropriate media with peptide was added. The freshly harvested cells were consistently >99% viable as assessed by their ability to exclude trypan blue. After stimulation with peptide, lysates were collected by lysing the cells in RIPA buffer in the presence of various phosphatase- and kinase-inhibitors. Protein content was analyzed and approximately 30 μg of each sample was loaded in a 12% SDS-PAGE gel. The gels were blotted onto nitrocellulose, blocked for 1 hour with 5% skim milk powder in Tris buffered saline (TBS) with 1% TritonX100. Phosphorylation was detected with phosphorylation-specific antibodies.


The results of peptide-induced phosphorylation are summarized in Table 46. SEQ ID NO: 2 was found to cause dose dependent phosphorylation of p38 and ERK1/2 in the mouse macrophage RAW cell line and the HBE cells. SEQ ID NO: 3 caused phosphorylation of MAP kinases in THP-1 human monocyte cell line and phosphorylation of ERK1/2 in the mouse RAW cell line.

TABLE 47Phosphorylation of MAP kinases in response to peptides.MAP kinasephosphorylatedCell LinePeptidep38ERK1/2RAW 264.7SEQ ID NO: 3+SEQ ID NO: 2++HBESEQ ID NO: 3+SEQ ID NO: 2++THP-1SEQ ID NO: 3++SEQ ID NO: 2









TABLE 48










Peptide Phosphorylation of MAP kinases in human blood monocytes.


SEQ ID NO: 1 at 50 μg/ml) was used to promote phosphorylation.












p38 phosphorylation

ERK1/2 phosphorylation













15 minutes
60 minutes
15 minutes
60 minutes







+

+
+










EXAMPLE 8
Cationic Peptides Protect Against Bacterial Infection by Enhancing the Immune Response

BALB/c mice were given 1×105 Salmonella and cationic peptide (200 μg) by intraperitoneal injection. The mice were monitored for 24 hours at which point they were euthanized, the spleen removed, homogenized and resuspended in PBS and plated on Luria Broth agar plates with Kanamycin (50 μg/ml). The plates were incubated overnight at 37° C. and counted for viable bacteria (Table 49 and 50). CD-1 mice were given 1×108 S. aureus in 5% porcine mucin and cationic peptide (200 μg) by intraperitoneal injection (Table 51). The mice were monitored for 3 days at which point they were euthanized, blood removed and plated for viable counts. CD-1 male mice were given 5.8×106 CFU EHEC bacteria and cationic peptide (200 μg) by intraperitoneal (IP) injection and monitored for 3 days (Table 52). In each of these animal models a subset of the peptides demonstrated protection against infections. The most protective peptides in the Salmonella model demonstrated an ability to induce a common subset of genes in epithelial cells (Table 53) when comparing the protection assay results in Tables 50 and 51 to the gene expression results in Tables 31-37. This clearly indicates that there is a pattern of gene expression that is consistent with the ability of a peptide to demonstrate protection. Many of the cationic peptides were shown not to be directly antimicrobial as tested by the Minimum Inhibitory Concentration (MIC) assay (Table 54). This demonstrates that the ability of peptides to protect against infection relies on the ability of the peptide to stimulate host innate immunity rather than on direct antimicrobial activity.

TABLE 49Effect of Cationic Peptides on Salmonella Infection in BALB/c mice.PeptideViable Bacteria in the SpleenStatistical SignificanceTreatment(CFU/ml)(p value)Control2.70 ± 0.84 × 105SEQ ID NO: 11.50 ± 0.26 × 1050.12SEQ ID NO: 62.57 ± 0.72 × 1040.03SEQ ID NO: 133.80 ± 0.97 × 1040.04SEQ ID NO: 174.79 ± 1.27 × 1040.04SEQ ID NO: 271.01 ± 0.26 × 1050.06
The BALB/c mice were injected IP with Salmonella and Peptide, and 24 h later the animals were euthanized, the spleen removed, homogenized, diluted in PBS and plate counts were done to determine bacteria viability.









TABLE 50










Effect of Cationic Peptides on Salmonella Infection in BALB/c mice.










Peptide Treatment
Viable Bacteria in the Spleen (CFU/ml)







Control
1.88 ± 0.16 × 104



SEQ ID NO: 48
1.98 ± 0.18 × 104



SEQ ID NO: 26
 7.1 ± 1.37 × 104



SEQ ID NO: 30
5.79 ± 0.43 × 103



SEQ ID NO: 37
1.57 ± 0.44 × 104



SEQ ID NO: 5
2.75 ± 0.59 × 104



SEQ ID NO: 7
 5.4 ± 0.28 × 103



SEQ ID NO: 9
1.23 ± 0.87 × 104



SEQ ID NO: 14
2.11 ± 0.23 × 103



SEQ ID NO: 20
2.78 ± 0.22 × 104



SEQ ID NO: 23
6.16 ± 0.32 × 104









The BALB/c mice were injected intraperitoneally with Salmonella and Peptide, and 24 h later the animals were euthanized, the spleen removed, homogenized, diluted in PBS and plate counts were done to determine bacteria viability.














TABLE 51










Effect of Cationic Peptides in a Murine S. aureus infection model.











# Mice Survived (3 days)/Total


Treatment
CFU/ml (blood)
mice in group





No Peptide
7.61 ± 1.7 × 103
6/8


SEQ ID NO: 1
0
4/4


SEQ ID NO: 27
2.25 ± 0.1 × 102
3/4


SEQ ID NO: 30
1.29 ± 0.04 × 102
4/4


SEQ ID NO: 37
9.65 ± 0.41 × 102
4/4


SEQ ID NO: 5
3.28 ± 1.7 × 103
4/4


SEQ ID NO: 6
1.98 ± 0.05 × 102
3/4


SEQ ID NO: 7
 3.8 ± 0.24 × 103
4/4


SEQ ID NO: 9
2.97 ± 0.25 × 102
4/4


SEQ ID NO: 13
4.83 ± 0.92 × 103
3/4


SEQ ID NO: 17
 9.6 ± 0.41 × 102
4/4


SEQ ID NO: 20
3.41 ± 1.6 × 103
4/4


SEQ ID NO: 23
4.39 ± 2.0 × 103
4/4







CD-1 mice were given 1 × 108 bacteria in 5% porcine mucin via intraperitoneal (IP) injection. Cationic peptide (200 μg) was given via a separate IP injection. The mice were monitored for 3 days at which point they were euthanized, blood removed and plated for viable counts. The following peptides were not effective in controlling S. aureus infection: SEQ ID NO: 48, SEQ ID NO: 26.














TABLE 52










Effect of Peptide in a Murine EHEC infection model.











Treatment
Peptide
Survival (%)















control
none
25



SEQ ID NO: 23
200 μg
100









CD-1 male mice (5 weeks old) were given 5.8 × 106 CFU EHEC bacteria via intraperitoneal (IP) injection. Cationic peptide (200 μg) was given via a separate IP injection. The mice were monitored for 3 days.














TABLE 53










Up-regulation of patterns of gene expression in A549 epithelial cells induced by


peptides that are active in vivo.









Fold Up regulation of Gene Expression



relative to Untreated Cells













Unstimulated
SEQ ID
SEQ ID
SEQ ID
SEQ ID


Target (Accession number)
Cell Intensity
NO: 30
NO: 7
NO: 13
NO: 37















Zinc finger protein (AF061261)
13
2.6
9.4
9.4
1.0


Cell cycle gene (S70622)
1.62
8.5
3.2
3.2
0.7


IL-10 Receptor (U00672)
0.2
2.6
9
4.3
0.5


Transferase (AF038664)
0.09
12.3
9.7
9.7
0.1


Homeobox protein (AC004774)
0.38
3.2
2.5
2.5
1.7


Forkhead protein (AF042832)
0.17
14.1
3.5
3.5
0.9


Unknown (AL096803)
0.12
4.8
4.3
4.3
0.6


KIAA0284 Protein (AB006622)
0.47
3.4
2.1
2.1
1.3


Hypothetical Protein (AL022393)
0.12
4.4
4.0
4.0
0.4


Receptor (AF112461)
0.16
2.4
10.0
10.0
1.9


Hypothetical Protein (AK002104)
0.51
4.7
2.6
2.6
1.0


Protein (AL050261)
0.26
3.3
2.8
2.8
1.0


Polypeptide (AF105424)
0.26
2.5
5.3
5.3
1.0


SPR1 protein (AB031480)
0.73
3.0
2.7
2.7
1.3


Dehydrogenase (D17793)
4.38
2.3
2.2
2.2
0.9


Transferase (M63509)
0.55
2.7
2.1
2.1
1.0


Peroxisome factor (AB013818)
0.37
3.4
2.9
2.9
1.4







The peptides SEQ ID NO: 30, SEQ ID NO: 7 and SEQ ID NO: 13 at concentrations of 50 μg/ml were each shown to increase the expression of a pattern of genes after 4 h treatment.





Peptide was incubated with the human A549 epithelial cells for 4 h and the RNA was isolated, converted into labelled cDNA probes and hybridised to Human Operon arrays (PRHU04).





The intensity of polynucleotides in control, unstimulated cells are shown in the second columns for labelling of cDNA (average of Cy3 and Cy5).





The Fold Up regulation column refers to the intensity of polynucleotide expression in peptide-simulated cells divided by the intensity of unstimulated cells.





The SEQ ID NO: 37 peptide was included as a negative control that was not active in the murine infection models.
















TABLE 54













MIC (μg/ml)













Peptide

E. coli


S. aureus


P. aerug.


S. typhim.


C. rhod.

EHEC
















Polymyxin
0.25
16
0.25
0.5
0.25
0.5


Gentamicin
0.25
0.25
0.25
0.25
0.25
0.5


SEQ ID NO: 1
32
>
96
64
8
4


SEQ ID NO: 5
128
>
>
>
64
64


SEQ ID NO: 6
128
>
>
128
64
64


SEQ ID NO: 7
>
>
>
>
>
>


SEQ ID NO: 8
>
>
>
>
>
>


SEQ ID NO: 9
>
>
>
>
>
>


SEQ ID NO: 10
>
>
>
>
>
64


SEQ ID NO: 12
>
>
>
>
>
>


SEQ ID NO: 13
>
>
>
>
>
>


SEQ ID NO: 14
>
>
>
>
>
>


SEQ ID NO: 15
128
>
>
>
128
64


SEQ ID NO: 16
>
>
>
>
>
>


SEQ ID NO: 17
>
>
>
>
>
>


SEQ ID NO: 19
8
16
16
64
4
4


SEQ ID NO: 2
4
16
32
16
64


SEQ ID NO: 20
8
8
8
8
16
8


SEQ ID NO: 21
64
64
96
64
32
32


SEQ ID NO: 22
8
12
24
8
4
4


SEQ ID NO: 23
4
8
8
16
4
4


SEQ ID NO: 24
16
16
4
16
16
4


SEQ ID NO: 26
0.5
32
64
2
2
0.5


SEQ ID NO: 27
8
64
64
16
2
4


SEQ ID NO: 28
>
>
>
64
64
128


SEQ ID NO: 29
2
>
>
16
32
4


SEQ ID NO: 30
16
>
128
16
16
4


SEQ ID NO: 31
>
>
128
>
>
64


SEQ ID NO: 33
16
32
>
16
64
8


SEQ ID NO: 34
8
>
>
32
64
8


SEQ ID NO: 35
4
128
64
8
8
4


SEQ ID NO: 36
32
>
>
32
32
16


SEQ ID NO: 37
>
>
>
>
>
>


SEQ ID NO: 38
0.5
32
64
4
8
4


SEQ ID NO: 40
4
32
8
4
4
2


SEQ ID NO: 41
4
64
8
8
2
2


SEQ ID NO: 42
1.5
64
4
2
2
1


SEQ ID NO: 43
8
128
16
16
8
4


SEQ ID NO: 44
8
>
128
128
64
64


SEQ ID NO: 45
8
>
128
128
16
16


SEQ ID NO: 47
4
>
16
16
4
4


SEQ ID NO: 48
16
>
128
16
1
2


SEQ ID NO: 49
4
>
16
8
4
4


SEQ ID NO: 50
8
>
16
16
16
8


SEQ ID NO: 51
4
>
8
32
4
8


SEQ ID NO: 52
8
>
32
8
2
2


SEQ ID NO: 53
4
>
8
8
16
8


SEQ ID NO: 54
64
>
16
64
16
32







Most cationic peptides studied here and especially the cationic peptides effective in infection models are not significantly antimicrobial. A dilution series of peptide was incubated with the indicated bacteria overnight in a 96-well plate. The lowest concentration of peptide that killed the bacteria was used as the MIC.





The symbol > indicates the MIC is too large to measure. An MIC of 4 μg/ml or less was considered clinically meaningful activity.





Abbreviations:






E. coli, Escherichia coli;






S. aureus, Staphylococcus aureus;






P. aerug, Pseudomonas aeruginosa;






S. Typhim, Salmonella enteritidis ssp. typhimurium;






C. rhod, Citobacter rhodensis;






EHEC, Enterohaemorrhagic E. coli.







EXAMPLE 9
Use of Polynucleotides Induced by Bacterial Signaling Molecules in Diagnostic/Screening


S. typhimurium LPS and E. coli 0111:B4 LPS were purchased from Sigma Chemical Co. (St. Louis, Mo.). LTA (Sigma) from S. aureus, was resuspended in endotoxin free water (Sigma). The Limulus amoebocyte lysate assay (Sigma) was performed on LTA preparations to confirm that lots were not significantly contaminated by endotoxin (i.e. <1 ng/ml, a concentration that did not cause significant cytokine production in the RAW cell assay). The CpG oligodeoxynucleotides were synthesized with an Applied Biosystems Inc., Model 392 DNA/RNA Synthesizer, Mississauga, ON., then purified and resuspended in endotoxin-free water (Sigma). The following sequences were used CpG: 5′-TCATGACGTTCCTGACGTT-3′ (SEQ ID NO: 57) and nonCpG: 5′-TTCAGGACTTTCCTCAGGTT-3′ (SEQ ID NO: 58). The nonCpG oligo was tested for its ability to stimulate production of cytokines and was found to cause no significant production of TNF-α or IL-6 and therefore was considered as a negative control. RNA was isolated from RAW 264.7 cells that had been incubated for 4 h with medium alone, 100 ng/ml S. typhimurium LPS, 1 μg/ml S. aureus LTA, or 1 μM CpG (concentrations that led to optimal induction of tumor necrosis factor (TNF-α) in RAW cells). The RNA was used to polynucleotiderate cDNA probes that were hybridized to Clontech Atlas polynucleotide array filters, as described above. The hybridization of the cDNA probes to each immobilized DNA was visualized by autoradiography and quantified using a phosphorimager. Results from at least 2 to 3 independent experiments are summarized in Tables 55-59. It was found that LPS treatment of RAW 264.7 cells resulted in increased expression of more than 60 polynucleotides including polynucleotides encoding inflammatory proteins such as IL-1β, inducible nitric oxide synthase (iNOS), MIP-1α, MIP-1β, MIP-2α, CD40, and a variety of transcription factors. When the changes in polynucleotide expression induced by LPS, LTA, and CpG DNA were compared, it was found that all three of these bacterial products increased the expression of pro-inflammatory polynucleotides such as iNOS, MIP-1α, MIP-2α, IL-1β, IL-15, TNFR1 and NF-κB to a similar extent (Table 57). Table 57 describes 19 polynucleotides that were up-regulated by the bacterial products to similar extents in that their stimulation ratios differed by less than 1.5 fold between the three bacterial products. There were also several polynucleotides that were down-regulated by LPS, LTA and CpG to a similar extent. It was also found that there were a number of polynucleotides that were differentially regulated in response to the three bacterial products (Table 58), which includes many of these polynucleotides that differed in expression levels by more than 1.5 fold between one or more bacterial products). LTA treatment differentially influenced expression of the largest subset of polynucleotides compared to LPS or CpG, including hyperstimulation of expression of Jun-D, Jun-B, Elk-1 and cyclins G2 and A1. There were only a few polynucleotides whose expression was altered more by LPS or CpG treatment. Polynucleotides that had preferentially increased expression due to LPS treatment compared to LTA or CpG treatment included the cAMP response element DNA-binding protein 1 (CRE-BPI), interferon inducible protein 1 and CACCC Box-binding protein BKLF. Polynucleotides that had preferentially increased expression after CpG treatment compared to LPS or LTA treatment included leukemia inhibitory factor (LIF) and protease nexin 1 (PN-1). These results indicate that although LPS, LTA, and CpG DNA stimulate largely overlapping polynucleotide expression responses, they also exhibit differential abilities to regulate certain subsets of polynucleotides.


The other polynucleotide arrays used are the Human Operon arrays (identification number for the genome is PRHU04-S1), which consist of about 14,000 human oligos spotted in duplicate. Probes were prepared from 5 μg of total RNA and labeled with Cy3 or Cy5 labeled dUTP. In these experiments, A549 epithelial cells were plated in 100 mm tissue culture dishes at 2.5×106 cells/dish, incubated overnight and then stimulated with 100 ng/ml E. coli O111:B4 LPS for 4 h. Total RNA was isolated using RNAqueous (Ambion). DNA contamination was removed with DNA-free kit (Ambion). The probes prepared from total RNA were purified and hybridized to printed glass slides overnight at 42° C. and washed. After washing, the image was. captured using a Perkin Elmer array scanner. The image processing software (Imapolynucleotide 5.0, Marina Del Rey, Calif.) determines the spot mean intensity, median intensities, and background intensities. An “in house” program was used to remove background. The program calculates the bottom 10% intensity for each subgrid and subtracts this for each grid. Analysis was performed with Polynucleotidespring software (Redwood City, Calif.). The intensities for each spot were normalized by taking the median spot intensity value from the population of spot values within a slide and comparing this value to the values of all slides in the experiment. The relative changes seen with cells treated with LPS compared to control cells can be found in the Tables below. A number of previously unreported changes that would be useful in diagnosing infection are described in Table 60.


To confirm and assess the functional significance of these changes, the levels of selected mRNAs and proteins were assessed and quantified by densitometry. Northern blots using a CD14, vimentin, and tristetraprolin-specific probe confirmed similar expression after stimulation with all 3 bacterial products (Table 60). Similarly measurement of the enzymatic activity of nitric oxide synthetase, iNOS, using Griess reagent to assess levels of the inflammatory mediator NO, demonstrated comparable levels of NO produced after 24 h, consistent with the similar up-regulation of iNOS expression (Table 59). Western blot analysis confirmed the preferential stimulation of leukaemia inhibitory factor (LIF, a member of the IL-6 family of cytokines) by CpG (Table 59). Other confirmatory experiments demonstrated that LPS up-regulated the expression of TNF-α and IL-6 as assessed by ELISA, and the up-regulated expression of MIP-2α, and IL-1β mRNA and down-regulation of DP-1 and cyclin D mRNA as assessed by Northern blot analysis. The analysis was expanded to a more clinically relevant ex vivo system, by examining the ability of the bacterial elements to stimulate pro-inflammatory cytokine production in whole human blood. It was found that E. coli LPS, S. typhimurium LPS, and S. aureus LTA all stimulated similar amounts of serum TNF-α, and IL-1β. CpG also stimulated production of these cytokines, albeit to much lower levels, confirming in part the cell line data.

TABLE 55Polynucleotides Up-regulated by E. coli O111:B4 LPSin A549 Epithelial Cells.Control:MediaRatio:AccessiononlyLPS/NumberGeneIntensitycontrolD87451ring finger protein 10715.8183.7AF061261C3H-type zinc finger protein565.936.7D17793aldo-keto reductase family 1,220.135.9member C3M14630prothymosin, alpha168.231.3AL049975Unknown145.662.3L04510ADP-ribosylation factor139.9213.6domain protein 1, 64 kDU10991G2 protein101.7170.3U39067eukaryotic translation61.015.9initiation factor 3, subunit 2X03342ribosomal protein L3252.610.5NM_004850Rho-associated, coiled-coil48.111.8containing protein kinase 2AK000942Unknown46.98.4AB040057serine/threonine protein42.144.3kinase MASKAB020719KIAA0912 protein41.89.4AB007856FEM-1-like death receptor41.215.7binding proteinJ02783procollagen-proline, 2-36.114.1oxoglutarate 4-dioxygenaseAL137376Unknown32.517.3AL137730Unknown29.411.9D25328phosphofructokinase, platelet27.38.5AF047470malate dehydrogenase 2,25.28.2NADM86752stress-induced-22.95.9phosphoprotein 1M90696cathepsin S19.66.8AK001143Unknown19.16.4AF038406NADH dehydrogenase17.771.5AK000315hypothetical protein17.317.4FLJ20308M54915pim-1 oncogene16.011.4D29011proteasome subunit, beta15.341.1type, 5AK000237membrane protein of15.19.4cholinergic synaptic vesiclesAL034348Unknown15.115.8AL161991Unknown14.28.1AL049250Unknown12.75.6AL050361PTD017 protein12.613.0U74324RAB interacting factor12.35.2M22538NADH dehydrogenase12.37.6D87076KIAA0239 protein11.66.5NM_006327translocase of inner11.510.0mitochondrial membrane 23(yeast) homologAK001083Unknown11.18.6AJ001403mucin 5, subtype B,10.853.4tracheobronchialM64788RAP1, GTPase activating10.77.6protein 1X06614retinoic acid receptor, alpha10.75.5U85611calcium and integring binding10.38.1proteinU23942cytochrome P450, 5110.110.2AL031983Unknown9.7302.8NM_007171protein-O-9.56.5mannosyltransferase 1AK000403hypothetical protein9.566.6FLJ20396NM_002950ribophorin I9.335.7L05515cAMP response element-8.96.2binding protein CRE-BPaX83368phosphoinositide-3-kinase,8.727.1catalytic, gamma polypeptideM30269nidogen (enactin)8.75.5M91083chromosome 11 open reading8.26.6frame 13D29833salivary proline-rich protein7.75.8AB024536immunoglobulin superfamily7.68.0containing leucine-rich repeatU39400chromosome 11 open reading7.47.3frame 4AF028789unc119 (C. elegans) homolog7.427.0NM_003144signal sequence receptor,7.35.9alpha (translocon-associatedprotein alpha)X52195arachidonate 5-lipoxygenase-7.313.1activating proteinU43895human growth factor-6.96.9regulated tyrosine kinasesubstrateL25876cyclin-dependent kinase6.710.3inhibitor 3L04490NADH dehydrogenase6.611.1Z18948S100 calcium-binding protein6.311.0D10522myristoylated alanine-rich6.15.8protein kinase C substrateNM_014442sialic acid binding Ig-like6.17.6lectin 8U81375solute carrier family 296.06.4AF041410malignancy-associated5.95.3proteinU24077killer cell immunoglobulin-5.814.4like receptorAL137614hypothetical protein4.86.8NM_002406mannosyl (alpha-1,3-)-4.75.3glycoprotein beta-1,2-N-acetylglucosaminyltransferaseAB002348KIAA0350 protein4.77.6AF165217tropomodulin 4 (muscle)4.612.3Z14093branched chain keto acid4.65.4dehydrogenase E1, alphapolypeptideU82671caltractin3.844.5AL050136Unknown3.65.0NM_005135solute carrier family 123.65.0AK001961hypothetical protein3.65.9FLJ11099AL034410Unknown3.221.3S74728antiquitin 13.19.2AL049714ribosomal protein L343.019.5pseudogene 2NM_014075PRO0593 protein2.911.5AF189279phospholipase A2, group IIE2.837.8J03925integrin, alpha M2.79.9NM_012177F-box protein Fbx52.626.2NM_004519potassium voltage-gated2.621.1channel, KQT-like subfamily,member 3M28825CD1A antigen, a polypeptide2.616.8X16940actin, gamma 2, smooth2.411.8muscle, entericX03066major histocompatibility2.236.5complex, class II, DO betaAK001237hypothetical protein2.118.4FLJ10375AB028971KIAA1048 protein2.09.4AL137665Unknown2.07.3
E. coli O111:B4 LPS (100 ng/ml) increased the expression of many polynucleotides in A549 cells as studied by polynucleotide microarrays. LPS was incubated with the A549 cells for 4 h and the RNA was isolated. 5 μg total RNA was used to make Cy3/Cy5 labelled cDNA probes and hybridised onto Human Operon arrays (PRHU04). The intensity of unstimulated cells is shown in the second column of Table 55. The “Ratio: LPS/control” column refers to the intensity of polynucleotide
# expression in LPS simulated cells divided by in the intensity of unstimulated cells.









TABLE 56










Polynucleotides Down-regulated by E. coli O111:B4 LPS in A549 Epithelial Cells.












Control:Media



Accession

only
Ratio:


Number
Gene
Intensity
LPS/control













NM_017433
myosin IIIA
167.8
0.03


X60484
H4 histone family member E
36.2
0.04


X60483
H4 histone family member D
36.9
0.05


AF151079
hypothetical protein
602.8
0.05


M96843
inhibitor of DNA binding 2, dominant
30.7
0.05



negative helix-loop-helix protein


S79854
deiodinase, iodothyronine, type III
39.4
0.06


AB018266
matrin 3
15.7
0.08


M33374
NADH dehydrogenase
107.8
0.09


AF005220
Homo sapiens mRNA for NUP98-HOXD13
105.2
0.09



fusion protein, partial cds


Z80783
H2B histone family, member L
20.5
0.10


Z46261
H3 histone family, member A
9.7
0.12


Z80780
H2B histone family, member H
35.3
0.12


U33931
erythrocyte membrane protein band 7.2
18.9
0.13



(stomatin)


M60750
H2B histone family, member A
35.8
0.14


Z83738
H2B histone family, member E
19.3
0.15


Y14690
collagen, type V, alpha 2
7.5
0.15


M30938
X-ray repair complementing defective
11.3
0.16



repair in Chinese hamster cells 5


L36055
eukaryotic translation initiation factor 4E
182.5
0.16



binding protein 1


Z80779
H2B histone family, member G
54.3
0.16


AF226869
5(3)-deoxyribonucleotidase; RB-associated
7.1
0.18



KRAB repressor


D50924
KIAA0134 gene product
91.0
0.18


AL133415
vimentin
78.1
0.19


AL050179
tropomyosin 1 (alpha)
41.6
0.19


AJ005579
RD element
5.4
0.19


M80899
AHNAK nucleoprotein
11.6
0.19


NM_004873
BCL2-associated athanogene 5
6.2
0.19


X57138
H2A histone family, member N
58.3
0.20


AF081281
lysophospholipase I
7.2
0.22


U96759
von Hippel-Lindau binding protein 1
6.6
0.22


U85977
Human ribosomal protein L12 pseudogene,
342.6
0.22



partial cds


D13315
glyoxalase I
7.5
0.22


AC003007
Unknown
218.2
0.22


AB032980
RU2S
246.6
0.22


U40282
integrin-linked kinase
10.1
0.22


U81984
endothelial PAS domain protein 1
4.7
0.23


X91788
chloride channel, nucleotide-sensitive, 1A
9.6
0.23


AF018081
collagen, type XVIII, alpha 1
6.9
0.24


L31881
nuclear factor I/X (CCAAT-binding
13.6
0.24



transcription factor)


X61123
B-cell translocation gene 1, anti-
5.3
0.24



proliferative


L32976
mitogen-activated protein kinase kinase
6.3
0.24



kinase 11


M27749
immunoglobulin lambda-like polypeptide 3
5.5
0.24


X57128
H3 histone family, member C
9.0
0.25


X80907
phosphoinositide-3-kinase, regulatory
5.8
0.25



subunit, polypeptide 2


Z34282
H. sapiens (MAR11) MUC5AC mRNA for
100.6
0.26



mucin (partial)


X00089
H2A histone family, member M
4.7
0.26


AL035252
CD39-like 2
4.6
0.26


X95289
PERB11 family member in MHC class I
27.5
0.26



region


AJ001340
U3 snoRNP-associated 55-kDa protein
4.0
0.26


NM_014161
HSPC071 protein
10.6
0.27


U60873
Unknown
6.4
0.27


X91247
thioredoxin reductase 1
84.4
0.27


AK001284
hypothetical protein FLJ10422
4.2
0.27


U90840
synovial sarcoma, X breakpoint 3
6.6
0.27


X53777
ribosomal protein L17
39.9
0.27


AL035067
Unknown
10.0
0.28


AL117665
DKFZP586M1824 protein
3.9
0.28


L14561
ATPase, Ca++ transporting, plasma
5.3
0.28



membrane 1


L19779
H2A histone family, member 0
30.6
0.28


AL049782
Unknown
285.3
0.28


X00734
tubulin, beta, 5
39.7
0.29


AK001761
retinoic acid induced 3
23.7
0.29


U72661
ninjurin 1
4.4
0.29


S48220
deiodinase, iodothyronine, type I
1,296.1
0.29


AF025304
EphB2
4.5
0.30


S82198
chymotrypsin C
4.1
0.30


Z80782
H2B histone family, member K
31.9
0.30


X68194
synaptophysin-like protein
7.9
0.30


AB028869
Unknown
4.2
0.30


AK000761
Unknown
4.3
0.30








E. coli O111:B4 LPS (100 ng/ml) decreased the expression of many polynucleotides in A549 cells as studied by polynucleotide microarrays. LPS was incubated with the A549 cells for 4 h and the RNA was isolated. 5 μg total RNA was used to make Cy3/Cy5 labeled cDNA probes and hybridized onto Human Operon arrays (PRHU04). The intensity of unstimulated cells is shown in the second column of the Table. The “Ratio: LPS/control” column refers to the intensity of polynucleotide



# expression in LPS simulated cells divided by in the intensity of unstimulated cells.













TABLE 57










Polynucleotides expressed to similar extents after stimulation by the bacterial


products LPS, LTA, and CpG DNA.













Control






Accession
Unstim.
Ratio
Ratio
Ratio


number
Intensity
LPS:Control
LTA:Control
CpG:Control
Protein/polynucleotide















M15131
20
82
80
55
IL-1β


M57422
20
77
64
90
tristetraprolin


X53798
20
73
77
78
MIP-2α


M35590
188
50
48
58
MIP-1β


L28095
20
49
57
50
ICE


M87039
20
37
38
45
iNOS


X57413
20
34
40
28
TGFβ


X15842
20
20
21
15
c-rel proto-oncopolynucleotide


X12531
489
19
20
26
MIP-1α


U14332
20
14
15
12
IL-15


M59378
580
10
13
11
TNFR1


U37522
151
6
6
6
TRAIL


M57999
172
3.8
3.5
3.4
NF-κB


U36277
402
3.2
3.5
2.7
I-κB (alpha subunit)


X76850
194
3
3.8
2.5
MAPKAP-2


U06924
858
2.4
3
3.2
Stat 1


X14951
592
2
2
2
CD18


X60671
543
1.9
2.4
2.8
NF-2


M34510
5970
1.6
2
1.4
CD14


X51438
2702
1.3
2.2
2.0
vimentin


X68932
4455
0.5
0.7
0.5
c-Fms


Z21848
352
0.5
0.6
0.6
DNA polymerase


X70472
614
0.4
0.6
0.5
B-myb







Bacterial products (100 ng/ml S. typhimurium LPS, 1 μg/ml S. aureus LTA or 1 μM CpG) were shown to potently induce the expression of several polynucleotides. Peptide was incubated with the RAW cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Atlas arrays.





The intensity of control, unstimulated cells is shown in the second column.





The “Ratio LPS/LTA/CpG: Control” column refers to the intensity of polynucleotide expression in bacterial product-simulated cells divided by the intensity of unstimulated cells.














TABLE 58










Polynucleotides that were differentially regulated by the bacterial products


LPS, LTA, and CpG DNA.













Unstim.






Accession
Control
Ratio
Ratio
Ratio


number
Intensity
LPS:Contrl
LTA:Contrl
CpG:Contrl
Protein/polynucleotide















X72307
20
1.0
23
1.0
hepatocyte growth factor


L38847
20
1.0
21
1.0
hepatoma transmembrane kinase







ligand


L34169
393
0.3
3
0.5
thrombopoietin


J04113
289
1
4
3
Nur77


Z50013
20
7
21
5
H-ras proto-oncopolynucleotide


X84311
20
4
12
2
Cyclin A1


U95826
20
5
14
2
Cyclin G2


X87257
123
2
4
1
Elk-1


J05205
20
18
39
20
Jun-D


J03236
20
11
19
14
Jun-B


M83649
20
71
80
42
Fas 1 receptor


M83312
20
69
91
57
CD40L receptor


X52264
20
17
23
9
ICAM-1


M13945
573
2
3
2
Pim-1


U60530
193
2
3
3
Mad related protein


D10329
570
2
3
2
CD7


X06381
20
55
59
102
Leukemia inhibitory factor (LIF)


X70296
20
6.9
13
22
Protease nexin 1 (PN-1)


U36340
20
38
7
7
CACCC Box-binding protein







BKLF


S76657
20
11
6
7
CRE-BPI


U19119
272
10
4
4
interferon inducible protein 1







Bacterial products (100 ng/ml S. typhimurium LPS, 1 μg/ml S. aureus LTA or 1 μM CpG) were shown to potently induce the expression of several polynucleotides. Peptide was incubated with the RAW cells for 4 h and the RNA was isolated, converted into labeled cDNA probes and hybridized to Atlas arrays.





The intensity of control, unstimulated cells is shown in the second column.





The “Ratio LPS/LTA/CpG: Control” column refers to the intensity of polynucleotide expression in bacterial product-simulated cells divided by the intensity of unstimulated cells.














TABLE 59










Confirmation of Table 57 and 58 Array Data.









Relative levels











Product
Untreated
LPS
LTA
CpG





CD14a
1.0
2.2 ± 0.4
1.8 ± 0.2
1.5 ± 0.3


Vimentina
1.0
 1.2 ± 0.07
 1.5 ± 0.05
 1.3 ± 0.07


Tristetraprolina
1.0
5.5 ± 0.5
5.5 ± 1.5
9.5 ± 1.5


LIFb
1.0
2.8 ± 1.2
2.7 ± 0.6
5.1 ± 1.6


NOc
8 ± 1.5
 47 ± 2.5
20 ± 3 
 21 ± 1.5








aTotal RNA was isolated from unstimulated RAW macrophage cells and cells treated for 4 hr with 100 ng/ml S. typhimurium LPS, 1 μg/ml S. aureus LTA, 1 μM CpG DNA or media alone and Northern blots were performed the membrane was probed for GAPDH, CD14, vimentin, and tristetraprolin as described previously [Scott et al]. The hybridization



# intensities of the Northern blots were compared to GAPDH to look for inconsistencies in loading. These experiments were repeated at least three times and the data shown is the average relative levels of each condition compared to media (as measured by densitometry) ± standard error.





bRAW 264.7 cells were stimulated with 100 ng/ml S. typhimurium LPS, 1 μg/ml S. aureus LTA, 1 μM CpG DNA or media alone for 24 hours. Protein lysates were prepared, run on SDS PAGE gels and western blots were performed to detect LIF (R&D Systems). These experiments were repeated at least three times and the data shown is the relative levels of LIF



# compared to media (as measured by densitometry) ± standard error.





cSupernatant was collected from RAW macrophage cells treated with 100 ng/ml S. typhimurium LPS, 1 μg/ml S. aureus LTA, 1 μM CpG DNA, or media alone for 24 hours and tested for the amount of NO formed in the supernatant as estimated from the accumulation of the stable NO metabolite nitrite with the Griess reagent as described previously [Scott, et al].





The data shown is the average of three experiments ± standard error.














TABLE 60










Pattern of Gene expression in A549 Human Epithelial cells up-regulated


by bacterial signaling molecules (LPS).








Accession



Number
Gene





AL050337
interferon gamma receptor 1


U05875
interferon gamma receptor 2


NM_002310
leukemia inhibitory factor receptor


U92971
coagulation factor II (thrombin) receptor-like 2


Z29575
tumor necrosis factor receptor superfamily member 17


L31584
Chemokine receptor 7


J03925
cAMP response element-binding protein


M64788
RAP1, GTPase activating protein


NM_004850
Rho-associated kinase 2


D87451
ring finger protein 10


AL049975
Unknown


U39067
eukaryotic translation initiation factor 3, subunit 2


AK000942
Unknown


AB040057
serine/threonine protein kinase MASK


AB020719
KIAA0912 protein


AB007856
FEM-1-like death receptor binding protein


AL137376
Unknown


AL137730
Unknown


M90696
cathepsin S


AK001143
Unknown


AF038406
NADH dehydrogenase


AK000315
hypothetical protein FLJ20308


M54915
pim-1 oncogene


D29011
proteasome subunit, beta type, 5


AL034348
Unknown


D87076
KIAA0239 protein


AJ001403
mucin 5, subtype B, tracheobronchial


J03925
integrin, alpha M








E. coli O111:B4 LPS (100 ng/ml) increased the expression of many polynucleotides in A549 cells as studied by polynucleotide microarrays. LPS was incubated with the A549 cells for 4 h and the RNA was isolated. 5 μg total RNA was used to make Cy3/Cy5 labelled cDNA probes and hybridised onto Human Operon arrays (PRHU04). The examples of polynucleotide



# expression changes in LPS simulated cells represent a greater than 2-fold intensity level change of LPS treated cells from untreated cells.






EXAMPLE 10
Altering Signaling to Protect Against Bacterial Infections

The Salmonella Typhimurium strain SL1344 was obtained from the American Type Culture Collection (ATCC; Manassas, Va.) and grown in Luria-Bertani (LB) broth. For macrophage infections, 10 ml LB in a 125 mL flask was inoculated from a frozen glycerol stock and cultured overnight with shaking at 37° C. to stationary phase. RAW 264.7 cells (1×105 cells/well) were seeded in 24 well plates. Bacteria were diluted in culture medium to give a nominal multiplicity of infection (MOI) of approximately 100, bacteria were centrifuged onto the monolayer at 1000 rpm for 10 minutes to synchronize infection, and the infection was allowed to proceed for 20 min in a 37° C., 5% CO2 incubator. Cells were washed 3 times with PBS to remove extracellular bacteria and then incubated in DMEM+10% FBS containing 100 μg/ml gentamicin (Sigma, St. Louis, Mo.) to kill any remaining extracellular bacteria and prevent re-infection. After 2 h, the gentamicin concentration was lowered to 10 μg/ml and maintained throughout the assay. Cells were pretreated with inhibitors for 30 min prior to infection at the following concentrations: 50 μM PD 98059 (Calbiochem), 50 μM U 0126 (Promega), 2 mM diphenyliodonium (DPI), 250 μM acetovanillone (apocynin, Aldrich), 1 mM ascorbic acid (Sigma), 30 mM N-acetyl cysteine (Sigma), and 2 mM Ng-L-monomethyl arginine (L-NMMA, Molecular Probes) or 2 mM NG-D-monomethyl arginine (D-NMMA, Molecular Probes). Fresh inhibitors were added immediately after infection, at 2 h, and 6-8 h post-infection to ensure potency. Control cells were treated with equivalent volumes of dimethylsulfoxide (DMSO) per mL of media. Intracellular survival/replication of S. Typhimurium SL1344 was determined using the gentamicin-resistance assay, as previously described. Briefly, cells were washed twice with PBS to remove gentamicin, lysed with 1% Triton X-100/0.1% SDS in PBS at 2 h and 24 h post-infection, and numbers of intracellular bacteria calculated from colony counts on LB agar plates. Under these infection conditions, macrophages contained an average of 1 bacterium per cell as assessed by standard plate counts, which permitted analysis of macrophages at 24 h post-infection. Bacterial filiamnentation is related to bacterial stress. NADPH oxidase and iNOS can be activated by MEK/ERK signaling. The results (Table 61) clearly demonstrate that the alteration of cell signaling is a method whereby intracellular Salmonella infections can be resolved. Thus since bacteria to up-regulate multiple genes in human cells, this strategy of blocking signaling represents a general method of therapy against infection.

TABLE 61Effect of the Signaling Molecule MEK on Intracellular Bacteria in IFN-γ-primed RAW cells.TreatmentaEffectb0NoneMEK inhibitorDecrease bacterial filamentation (bacterial stress)cU 0126Increase in the number of intracellular S. TyphimuriumMEK inhibitorDecrease bacterial filamentation (bacterial stress)cPD 98059Increase in the number of intracellular S. TyphimuriumNADPHDecrease bacterial filamentation (bacterial stress)coxidase inhibitordIncrease in the number of intracellular S. Typhimurium


EXAMPLE 11
Anti-Viral Activity

SDF-1, a C-X-C chemokine is a natural ligand for HIV-1 coreceptor-CXCR4. The chemokine receptors CXCR4 and CCR5 are considered to be potential targets for the inhibition of HIV-1 replication. The crystal structure of SDF-1 exhibits antiparallel β-sheets and a positively charged surface, features that are critical in binding to the negatively charged extracellular loops of CXCR4. These findings suggest that chemokine derivatives, small-size CXCR4 antagonists, or agonists mimicking the structure or ionic property of chemokines may be useful agents for the treatment of X4 HIV-1 infection. It was found that the cationic peptides inhibited SDF-1 induced T-cell migration suggesting that the peptides may act as CXCR4 antagonists. The migration assays were performed as follows. Human Jurkat T cells were resuspended to 5×106/ml in chemotaxis medium (RPMI 1640/10 mM Hepes/0.5% BSA). Migration assays were performed in 24 well plates using 5 pm polycarbonate Transwell inserts (Costar). Briefly, peptide or controls were diluted in chemotaxis medium and placed in the lower chamber while 0.1 ml cells (5×106/ml) was added to the upper chamber. After 3 hr at 37° C., the number of cells that had migrated into the lower chamber was determined using flow cytometry. The medium from the lower chamber was passed through a FACscan for 30 seconds, gating on forward and side scatter to exclude cell debris. The number of live cells was compared to a “100% migration control” in which 5×105/ml cells had been pipetted directly into the lower chamber and then counted on the FACscan for 30 seconds. The results demonstrate that the addition of peptide results in an inhibition of the migration of Human Jurkat T-cells (Table 62) probably by influencing CXCR4 expression (Tables 63 and 64).

TABLE 62Peptide inhibits the migration of human Jurkat-T cells:Migration (%)PositiveSDF-1SDF-1 + SEQ 1DNegativeExperimentcontrol(100 ng/ml)1 (50 μg/ml)control1100%32%0%<0.01%2100%40%0%   0%









TABLE 63










Corresponding polynucleotide array data to Table 56:











Poly-






nucle-
Poly-


otide/
nucleotide
Unstimulated
Ratio
Accession


Protein
Function
Intensity
peptide:Unstimulated
Number





CXCR-
Chemokine
36
4
D87747


4
receptor
















TABLE 64










Corresponding FACs data to Tables 62 and 63:













Fold Increase in Protein




Concentration
Expression



Peptide
(μg/ml)
CXCR-4















SEQ ID NO: 1
10
No change



SEQ ID NO: 1
50
1.3 ± 0.03



SEQ ID NO: 1
100
1.6 ± 0.23



SEQ ID NO: 3
100
1.5 ± 0.2 










EXAMPLE 12
Synergistic Combinations

Methods and Materials



S. aureus was prepared in phosphate buffered solution (PBS) and 5% porcine mucin (Sigma) to a final expected concentration of 1-4×107 CFU/ml. 100 μl of S. aureus (mixed with 5% porcine, mucin) was injected intraperitoneally (IP) into each CD-1 mouse (6-8 weeks female weighing 20-25 g (Charles River)). Six hours after the onset of infection, 100 μl of the peptide was injected (50-200 μg total) IP along with 0.1 mg/kg Cefepime. After 24 hours, animals were sacrificed and heart puncture was performed to remove 100 μl of blood. The blood was diluted into 1 ml PBS containing Heparin. This was then ftrther diluted and plated for viable colony counts on Mueller-Hinton agar plates (10−1, 10−2, 10−3 & 10−4). Viable colonies, colony-forming units (CFU), were counted after 24 hours. Each experiment was carried out a minimum of three times. Data is presented as the average CFU+standard error per treatment group (8-10 mice/group).


Experiments were carried out with peptide and sub-optimal Cefepime given 6 hours after the onset of systemic S. aureus infection (FIG. 1). The data in FIG. 1 is presented as the mean±standard error of viable counts from blood taken from the mice 24 hrs after the onset of infection. The combination of sub optimal antibiotic (cefepime) dosing and SEQ ID NO: 7 resulted in improved therapeutic efficacy. The ability of the peptides to work in combination with sub-optimal concentrations of an antibiotic in a murine infection model is an important finding. It suggests the potential for extending the life of antibiotics in the clinic and reducing incidence of antibiotic resistance.


SEQ ID NO: 1, as an example, induced phosphorylation and activation of the mitogen activated protein kinases, ERK1/2 and p38 in human peripheral blood-derived monocytes and a human bronchial epithelial cell line but not in B- or T-lymphocytes. Phosphorylation was not dependent on the G-protein coupled receptor, FPRL-1, which was previously proposed to be the receptor for SEQ ID NO: 1-induced chemotaxis on human monocytes and T cells. Activation of ERK1/2 and p38 was markedly increased by the presence of granulocyte macrophage-colony stimulating factor (GM-CSF), but not macrophage-colony stimulating factor (M-CSF). Exposure to SEQ ID NO: 1 also led to the activation of Elk1/2, a transcription factor that is downstream of and activated by phosphorylated ERK1/2, as well as the up-regulation of various Elk-1 controlled genes. The ability of SEQ ID NO: 1 to signal through these pathways has broad implications in immunity, monocyte activation, proliferation and differentiation.


SEQ ID NO: 1 (sequence LGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES), was synthesized by Fmoc [(N-(9-fluorenyl) methoxycarbonyl)] chemistry at the Nucleic Acid/Protein Synthesis (NAPS) Unit at UBC. Human recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-4 (IL-4) and macrophage colony-stimulating factor (M-CSF) were purchased from Research Diagnostics Inc. (Flanders, N.J., USA). Pertussis toxin was supplied by List Biological Laboratories Inc. (Campbell, Calif., USA).


Blood monocytes were prepared using standard techniques. Briefly, 100 ml of fresh human venous blood was collected in sodium heparin Vacutainer collection tubes (Becton Dickinson, Mississauga, ON, Canada) from volunteers according to UBC Clinical Research Ethics Board protocol C02-0091. The blood was mixed, at a 1:1 ratio, with RPMI 1640 media [supplemented with 10% v/v fetal calf serum (FBS), 1% L-glutamine, 1 nM sodium pyruvate] in an E-toxa-clean (Sigma-Aldrich, Oakville, ON, Canada) washed, endotoxin-free bottle. PBMC were separated using Ficoll-Paque Plus (Amersham Pharmacia Biotech, Baie D'Urfé, PQ, Canada) at room temperature and washed with phosphate buffered saline (PBS). Monocytes were enriched with the removal of T-cells by rosetting with fresh sheep red blood cells (UBC animal care unit) pre-treated with Vibrio cholerae neuraminidase (Calbiochem Biosciences Inc., La Jolla, Calif., USA) and repeat separation by Ficoll Paque Plus. The enriched monocytes were washed with PBS, then cultured (approximately 2-3×106 per well) for 1 hour at 37° C. followed by the removal of non-adherent cells; monocytes were >95% pure as determined by flow cytometry (data not shown). B-lymphocytes were isolated by removing non-adherent cells and adding them to a new plate for one hour at 37° C. This was repeated a total of three times. Any remaining monocytes adhered to the plates, and residual non-adherent cells were primarily B cells. Cells were cultured in Falcon tissue culture 6-well plates (Becton Dickinson, Mississauga, ON, Canada). The adherent monocytes were cultured in 1 ml media at 37° C. in which SEQ ID NO: 1 and/or cytokines dissolved in endotoxin-free water (Sigma-Aldrich, Oakville, ON, Canada) were added. Endotoxin-free water was added as a vehicle control. For studies using pertussis toxin the media was replaced with 1 ml of fresh media containing 100 ng/ml of toxin and incubated for 60 min at 37° C. SEQ ID NO: 1 and cytokines were added directly to the media containing pertussis toxin. For the isolation of T lymphocytes, the rosetted T cells and sheep red blood cells were resuspended in 20 ml PBS and 10 ml of distilled water was added to lyse the latter. The cells were then centrifuged at 1000 rpm for 5 min after which the supernatant was removed. The pelleted T cells were promptly washed in PBS and increasing amounts of water were added until all sheep red blood cells had lysed. The remaining T cells were washed once in PBS, and viability was confirmed using a 0.4% Trypan blue solution. Primary human blood monocytes and T cells were cultured in RPMI 1640 supplemented with 10% v/v heat-inactivated FBS, 1% v/v L-glutamine, 1 nM sodium pyruvate (GIBCO Invitrogen Corporation, Burlington, ON, Canada). For each experiment between two and eight donors were used.


The simian virus 40-transformed, immortalized 16HBE4o-bronchial epithelial cell line was a generous gift of Dr. D. Gruenert (University of California, San Francisco, Calif.). Cells were routinely cultured to confluence in 100% humidity and 5% CO2 at 37° C. They were grown in Minimal Essential media with Earles' salts (GIBCO Invitrogen Corporation, Burlington, ON, Canada) containing 10% FBS (Hyclone), 2mM L-glutamine. For experiments, cells were grown on Costar Transwell inserts (3-μm pore size, Fischer Scientific) in 24-well plates. Cells were seeded at 5×104 cells per 0.25 ml of media on the top of the inserts while 0.95 ml of media was added to the bottom of the well and cultured at 37° C. and 5% CO2. Transmembrane resistance was measured daily with a Millipore voltohmeter and inserts were used for experiments typically after 8 to 10 days, when the resistance was 500-700 ohms. The cells were used between passages 8 and 20.


Western Immunoblotting—After stimulation, cells were washed with ice-cold PBS containing 1 mM vanadate (Sigma). Next 125 μl of RIPA buffer (50 mM Tris-HCI, pH 7.4, NP-40 1%, sodium deoxycholate 0.25%, NaCl 150 mM, EDTA 1 mM, PMSF 1 mM, Aprotinin, leupeptin, pepstatin 1 μg/ml each, sodium orthovanadate 1 mM, NaF 1 mM) was added and the cells were incubated on ice until they were completely lysed as assessed by visual inspection. The lysates were quantitated using a BCA assay (Pierce). 30 μg of lysate was loaded onto 1.5 mm thick gels, which were run at 100 volts for approximately 2 hours. Proteins were transferred to nitrocellulose filters for 75 min at 70 V. The filters were blocked for 2 hours at room temperature with 5% skim milk in TBST (10 mM Tris-HCI pH 8, 150 mM NaCl, 0.1% Tween-20). The filters were then incubated overnight at 4° C. with the anti-ERK1/2-P or anti-p38-P (Cell Signaling Technology, Ma) monoclonal antibodies. Immunoreactive bands were detected using horseradish peroxidase-conjugated sheep anti-mouse IgG antibodies (Amersham Pharmacia, New Jersey) and chemiluminescence detection (Sigma, Mo). To quantify bands, the films were scanned and then quantified by densitometry using the software program, ImageJ. The blots were reprobed with β-actin antibody (ICN Biomedical Incorporated, Ohio) and densitometry was performed to allow correction for protein loading.


Kinase Assay—An ERK1/2 activity assay was performed using a non-radioactive kit (Cell Signaling Technology). Briefly, cells were treated for 15 min and lysed in lysis buffer. Equal amounts of proteins were immunoprecipitated with an immobilized phospho-ERK1/2 antibody that reacts only with the phosphorylated (i.e. active) form of ERK1/2. The immobilized precipitated enzymes were then used for the kinase assay using Elk-1 followed by Western blot analysis with antibodies that allow detection and quantitation of phosphorylated substrates.


Quantification of IL-8—Human IL-8 from supernatants of 16HBE40-cells was measured by using the commercially available enzyme-linked immunosorbent assay kit (Biosource) according to the manufacturer's instructions.


Semiquantitative RT-PCR—Total RNA from two independent experiments was isolated from 16HBE4o-cells using RNaqueous (Ambion) as described by the manufacturer. The samples were DNase treated, and then cDNA synthesis was accomplished by using a first-strand cDNA synthesis kit (Gibco). The resultant cDNAs were used as a template in PCRs for various cytokine genes:

(SEQ ID NO: 59)MCP-15′-TCATAGCAGCCACCTTCATTC-3′;(SEQ ID NO: 60)5′-TAGCGCAGATTCTTGGGTTG-3′;(SEQ ID NO: 61)MCP-35′-TGTCCTTTCTCAGAGTGGTTCT-3′;(SEQ ID NO: 62)5′-TGCTTCCATAGGGACATCATA-3′(SEQ ID NO: 63)IL-65′-ACCTGAACCTTCCAAAGATGG-3′;(SEQ ID NO: 64)5′-GCGCAGAATGAGATGAGTTG-3′;and(SEQ ID NO: 65)IL-85′-GTGCAGAGGGTTGTGGAGAAG-3′;(SEQ ID NO: 66)5′-TTCTCCCGTGCAATATCTAGG-3′


Each RT-PCR reaction was performed in at least duplicate. Results were analysed in the linear phase of amplification and normalized to the housekeeping control, glyceraldehyde-3-phosphate dehydrogenase. Reactions were verified for RNA amplification by including controls without reverse transcriptase.


Peptides induce ERK1/2 and p38 phosphorylation in peripheral blood derived monocytes. To determine if peptide induced the activation of the MAP kinases, ERK1/2 and/or p38, peripheral blood derived monocytes were treated with 50 μg/ml SEQ ID NO: 1 or water (as a vehicle control) for 15 min. To visualize the activated (phosphorylated) form of the kinases, Western blots were performed with antibodies specific for the dually phosphorylated form of the kinases (phosphorylation on Thr202+Tyr204 and Thr180+Tyr182 for ERK1/2 and p38 respectively). The gels were re-probed with an antibody for β-actin to normalize for loading differences. In all, an increase in phosphorylation of ERK1/2 (n=8) and p38 (n=4) was observed in response to SEQ ID NO: 1 treatment (FIG. 2).



FIG. 2 shows exposure to SEQ ID NO: 1 induces phosphorylation of ERK1/2 and p38. Lysates from human peripheral blood derived monocytes were exposed to 50 μg/ml of SEQ ID NO: 1 for 15 minutes. A) Antibodies specific for the phosphorylated forms of ERK and p38 were used to detect activation of ERK1/2 and p38. All donors tested showed increased phosphorylation of ERK1/2 and p38 in response to SEQ ID NO: 1 treatment. One representative donor of eight. Relative amounts of phosphorylation of ERK (B) and p38(C) were determined by dividing the intensities of the phosphorylated bands by the intensity of the corresponding control band as described in the Materials and Methods.


Peptide induced activation of ERK1/2 is greater in human serum than in fetal bovine serum. It was demonstrated that SEQ ID NO: 1 induced phosphorylation of ERK1/2 did not occur in the absence of serum and the magnitude of phosphorylation was dependent upon the type of serum present such that activation of ERK1/2 was far superior in human serum (HS) than in fetal bovine serum (FBS).



FIG. 3 shows SEQ ID NO: 1 induced phosphorylation of ERK1/2 does not occur in the absence of serum and the magnitude of phosphorylation is dependent upon the type of serum present. Human blood derived monocytes were treated with 50 μg/ml of SEQ ID NO: 1 for 15 minutes. Lysates were run on a 12% acrylamide gel then transferred to nitrocellulose membrane and probed with antibodies specific for the phosphorylated (active) form of the kinase. To normalize for protein loading, the blots were reprobed with β-actin. Quantification was done with ImageJ software.The FIG. 3 inset demonstrates that SEQ ID NO: 1 is unable to induce MAPK activation in human monocytes under serum free conditions. Cells were exposed to 50 mg/ml of SEQ ID NO: 1 (+), or endotoxin free water (−) as a vehicle control, for 15 minutes. (A) After exposure to SEQ ID NO: 1 in media containing 10% fetal calf serun, phosphorylated ERK1/2 was detectable, however, no phosphorylation of ERK1/2 was detected in the absence of serum (n=3). (B) Elk-1, a transcription factor downstream of ERK1/2, was activated (phosphorylated) upon exposure to 50 μg/ml of SEQ ID NO: 1 in media containing 10% fetal calf serum, but not in the absence of serum (n=2).


Peptide induced activation of ERK1/2 and p38 is dose dependent and demonstrates synergy with GM-CSF. GM-CSF, IL-4, or M-CSF (each at 100 ng/ml) was added concurrently with SEQ ID NO: 1 and phosphorylation of ERK1/2 was measured in freshly isolated human blood monocytes. ERK1/2 phosphorylation was evident when cells were treated with 50 μg/ml of SEQ ID NO: 1 (8.3 fold increase over untreated, n=9) but not at lower concentrations (n=2). In the presence of 100 ng/ml GM-CSF, SEQ ID NO: 1-induced ERK1/2 phosphorylation increased markedly (58 fold greater than untreated, n=5). Furthermore, in the presence of GM-CSF, activation of ERK1/2 occurred in response to concentrations of 5 and 10 μg/ml of SEQ ID NO: 1, respectively, in the two donors tested (FIG. 4). This demonstrates that SEQ ID NO: 1 induced activation of ERK1/2 occurred at a lower threshold in the presence of GM-CSF, a cytokine found locally at sites of infection.



FIG. 4 shows SEQ ID NO: 1 induced activation of ERK1/2 occurs at lower concentrations and is amplified in the presence of certain cytokines. When freshly isolated monocytes were stimulated in media containing both GM-CSF (100 ng/ml) and IL-4 (100 ng/ml) SEQ ID NO: 1 induced phosphorylation of ERK1/2 was apparent at concentrations as low as 5 μg/ml. This synergistic activation of ERK1/2 seems to be due primarily to GM-CSF.


Activation of ERK1/2 leads to transcription of Elk-1 controlled genes and secretion of IL-8. IL-8 release is governed, at least in part, by activation of the ERK1/2 and p38 kinases. In order to determine if peptide could induce IL-8 secretion the human bronchial cell line, 16HBE4o-, was grown to confluency in Transwell filters, which allows for cellular polarization with the creation of distinct apical and basal surfaces. When the cells were stimulated with 50 μg/ml of SEQ ID NO: 1 on the apical surface for four hours a statistically significant increase in the amount of IL-8 released into the apical supernatant was detected (FIG. 5). To determine the downstream transcriptional effects of peptide-induced MAP kinase activation, the expression of genes known to be regulated by ERK1/2 or p38 was assessed by RT-PCR. RT-PCR was performed on RNA isolated from 16HBE4o-cells, treated for four hours with 50lg/ml of SEQ ID NO: 1 in the presence of serum, from two independent experiments. MCP-1 and IL-8 have been demonstrated to be under the transcriptional control of both ERK1/2 and p38, consistent with this they are up-regulated 2.4 and 4.3 fold respectively. Transcription of MCP-3 has not previously been demonstrated to be influenced by the activation of the mitogen activated protein kinases, consistent with this, expression is not affected by peptide treatment. (FIG. 5). These data are consistent with the hypothesis that activation of the activation of the ERK1/2 and p38 signaling pathways has functional effects on transcription of cytokine genes with immunomodulatory functions. The inset to FIG. 3B also demonstrates that peptide induced the phosphorylation of transcription facor Elk-1 in a serum dependent manner.



FIG. 5 shows peptide affects both transcription of various cytokine genes and release of IL-8 in the 16HBE4o-human bronchial epithelial cell line. Cells were grown to confluency on a semi-permeable membrane and stimulated on the apical surface with 50 μg/ml of SEQ ID NO: 1 for four hours. A) SEQ ID NO: 1 treated cells produced significantly more IL-8 than controls, as detected by ELISA in the supernatant collected from the apical surface, but not from the basolateral surface. Mean±SE of three independent experiments shown, asterisk indicates p=0.002.B) RNA was collected from the above experiments and RT-PCR was performed. A number of cytokine genes known to be regulated by either ERK1/2 or p38 were up-regulated upon stimulation with peptide. The average of two independent experiments is shown.


EXAMPLE 13
Modulation of an Inflammatory Response

The innate immune response is a dynamic system since it can be triggered by receptor. recognition of conserved bacterial components, initiating a broad inflammatory response to infectious agents, but must be able maintain homeostasis in the presence of commensal organisms, which contain many of these same conserved components. A delicate balance of pro- and anti-inflammatory mediators is vital for efficient functioning of the immune system under these disparate circumstances. In recent years, there has been speculation and some evidence implicating the sole human cathelicidin, SEQ ID NO: 1, in maintaining homeostasis, combating pathogenic challenge, and protecting against endotoxemia, an extreme inflammation-like condition (Devine D A, et al. Cationic peptides: distribution and mechanisms of resistance. Curr Pharm Des 2002; 8:703-14; Ciornei C D, et al. Antimicrobial and chemoattractant activity, Lipopolysaccharide neutralization, cytotoxicity, and inhibition by serum of analogs of human cathelicidin LL-37. Antimicrob Agents Chemother 2005; 49:2845-50). The data presented herein demonstrate that SEQ ID NO: 1 is an important component of human immunity that regulates the balance of pro- and anti-inflammatory molecules both under homeostatic conditions and during endotoxin challenge (i.e., infection situations).


Materials and Methods


Cell Isolation and Cell Lines—Human monocytic cells, THP-1 (Tsuchiya S, et al. Establishment and characterization of a human acute monocytic leukemia cell line (THP-1). Int J Cancer 1980; 26:171-6), were obtained from American type culture collection, ATCC® (TIB-202) and were grown in suspension in RPMI-1640 media (Gibco®, Invitrogen™ Life technologies, Burlington, ON), supplemented with 10% (v/v) heat inactivated fetal bovine serum (FBS), 2 mM L-glutamine and 1 mM sodium pyruvate (all from Invitrogen Life Technologies). Cultures were maintained at 37° C. in a humidified 5% (v/v) CO2 incubator up to a maximum of six passages. THP-1 cells at a density of 1×106 cells/ml were treated with 0.3 μg/ml phorbol 12-myristate 13-acetate (PMA; Sigma-Aldrich Canada, Oakville ON) for 24 hr (Tsuchiya S, et al. Induction of maturation in cultured human monocytic leukemia cells by a phorbol diester. Cancer Res 1982; 42:1530-6), inducing plastic-adherent cells that were further rested in complete RPMI-1640 medium for an additional 24 hr prior to stimulations with various treatments. Venous blood (20 ml) from healthy volunteers was collected in Vacutainer collection tubes containing sodium heparin as an anticoagulant (Becton Dickinson, Mississauga, ON) in accordance with UBC ethical approval and guidelines. Blood was diluted 1:1 with complete RPMI 1640 medium and separated by centrifugation over a Ficoll-Paqueg Plus (Amersham Biosciences, Piscataway, N.J., USA) density gradient. White blood cells were isolated from the buffy coat, washed twice in RPMI 1640 complete medium, and the number of peripheral blood mononuclear cells (PBMC) was determined by trypan blue exclusion. PBMC (5×105) were seeded into 12-well tissue culture dishes (Falcon; Becton Dickinson) at 1×106 cells/ml at 37° C. in 5% CO2. All experiments using human THP-1 cells or PBMCs involved at least three biological replicates.


Stimulants, Reagents and Antibodies—LPS was isolated from P. aeruginosa H103 using the Darveau-Hancock method as previously described (Darveau RP, et al. Procedure for isolation of bacterial lipopolysaccharides from both smooth and rough Pseudomonas aeruginosa and Salmonella typhimurium strains. J Bacteriol 1983; 155:831-8). Briefly, P. aeruginosa was grown overnight in LB broth at 37° C. Cells were collected and washed and the isolated LPS pellets were extracted with a 2:1 chloroform:methanol solution to remove contaminating lipids. Purified LPS samples were quantitated using an assay for the specific sugar 2-keto-3-deoxyoctosonic acid (KDO assay) and then resuspended in endotoxin-free water (Sigma-Aldrich).


TLR2 agonists lipoteichoic acid (LTA) from S. aureus and a synthetic tripalmitoylated lipopeptide, Pam3CSK4, were purchased from InvivoGen (San Diego, Calif., USA). TLR9 agonist CpG oligodeoxynucleotide #2007 (Krieg AM. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature 1995; 374:546-9) was a gift from Dr. Lorne Babuik (Vaccine and Infectious Disease org., SK, Canada). Recombinant human TNFα and recombinant human ILL1β were obtained from Research Diagnostics Inc., (Flanders, N.J., USA). All reagents were tested for endotoxin and reconstituted in endotoxin-free water. LTA from S. aureus used in this study had 1.25 EU of endotoxin/μg of LTA. Polymyxin B was purchased from InvivoGen, Actinomycin D (transcriptional inhibitor) was purchased from Calbiochem-Novabiochem Corporation (La Jolla, Calif.) and Monensin (inhibitor of protein secretion) was purchased from eBiosciences., CA, USA. A cationic peptide, SEQ ID NO: 1, was synthesized using F-moc chemistry at the Nucleic Acid/Protein Synthesis Unit, University of British Columbia (Vancouver, BC, Canada). The synthetic peptide was re-suspended in endotoxin-free water and stored at −20° C. until flurther use.


Rabbit polyclonal antibodies against the NFκB subunits p105/p50, p65 and RelB were purchased from Cell Signaling Technologies (Mississauga, ON, Canada). Rabbit polyclonal antibody against the NFκB subunit c-Rel was purchased from Chemicon International (Temecula, CA, USA) and mouse IgG2a monoclonal antibody against NFκB subunit p100/p52 was purchased from Upstate Cell Signaling Solutions (Lake Placid, N.Y., USA). HRP-conjugated goat anti-rabbit and anti-mouse IgG antibodies were purchased from Cell Signaling Technologies and Amersham Biosciences respectively.


Treatment with inflammatory stimuli, peptide or inhibitors—THP-1 cells or PBMC were stimulated with LPS (10 or 100 ng/ml), LTA (1 μg/ml), Pam3CSK4 (100 ng/ml), CpG-ODN 2007 (2 μg/ml), recombinant human TNFα (50 ng/ml) or recombinant human IL1β (50 ng/ml) for 1, 2, 4, or 24 hours. SEQ ID NO: 1 (0.5-50 lg/ml) was added simultaneously or 30 min after addition of the stimulants. Alternatively, cells were stimulated with SEQ ID NO: 1 (20 μg/ml) for 30 min, washed with RPMI complete media to remove the peptide and then stimulated with LPS (100 ng/ml). Polymyxin B (0.1 mg/ml), actinomycin D (4 μg/ml), or monensin (working concentration as per the manufacturer's instructions) were added to the THP-1 cells 30 min prior to stimulants.


Detection ofcytokines—Following incubation of the cells under various treatment regimens, the tissue culture supernatants were centrifuged at 1000× g for 5 min, then at 10,000×g for 2 min to obtain cell-free samples. Supernatants were aliquoted and then stored at −20° C. prior to assay for various cytokines. TNFα and IL-8 secretion were detected with a capture ELISA (eBioscience and BioSource International Inc., CA, USA respectively) using either tissue culture supernatants or the nuclear and cytoplasmic extracts (see below) as per the experimental design. All assays were performed in triplicate. The concentration of the cytokines in the culture medium was quantified by establishing a standard curve with serial dilutions of the recombinant human TNFα or IL-8 respectively. Alternatively, five cytokines (GM-CSF, IL-1β, IL-6, IL-8 and TNFα) were measured simultaneously using the Human Cytokine 5-Plex kit from Biosource International Inc., (Medicorp Inc., Montreal, Canada) as per the manufacturer's instructions. The multiplex bead immunoassays were analyzed using Luminex 100™ StarStation software (Applied Cytometry Systems, Sacramento, CA, USA).


RNA extraction, amplification and hybridization to DNA microarrays—RNA was isolated from THP-1 cells with RNeasy Mini kit, treated with RNase-Free DNase (Qiagen Inc., Canada) and eluted in RNase-free water (Ambion Inc., Austin, Tex., USA) as per the manufacturer's instructions. RNA concentration, integrity and purity were assessed by Agilent 2100 Bioanalyzer using RNA 6000 Nano kits (Agilent Technologies; USA). RNA was (reverse) transcribed with incorporation of amino-allyl-UTP (aa-UTP) using the MessageAmpII™ amplification kit, according to the manufacturer's instructions, then column purified and eluted in nuclease-free water. Column purified samples were labeled with mono-functional dyes, Cyanine-3 and Cyanine-5 (Amersham Biosciences), according to manufacturer's instructions, and then purified using the Mega Clear kit (Ambion). Yield and fluorophore incorporation was measured using Lambda 35 UV/VIS fluorimeter (PerkinElmer Life and Analytical Sciences, Inc., USA). Microarray slides were printed with the human genome 21K Array-Ready Oligo Set™ (Qiagen Inc., USA) at The Jack Bell Research Center (Vancouver, BC, Canada). The slides were pre-hybridized for 45 min at 48° C. in pre-hybridization buffer containing 5×SSC (Ambion), 0.1% (w/v) SDS and 0.2% (w/v) BSA. Equivalent (20 pmol) cyanine labeled samples from control and treated cells were then mixed and hybridized on the array slides, in Ambion SlideHyb™ buffer#2 (Ambion) for 18 hr at 37° C. in a hybridization oven. Following hybridization, the slides were washed twice in 1×SSC/0.1% sodium dodecyl sulphate (SDS) for 5 min at 65° C., then twice in 1×SSC and 0.1×SSC for 3 min each at 42° C. Slides were centrifugated for 5 min at 1000×g, dried and scanned using ScanArray™ Express software/scanner (scanner and software by Packard BioScience BioChip Technologies) and the images were quantified using ImaGene™ (BioDiscovery Inc., El Segundo, Calif., USA).


Analysis of DNA Microarrays—Assessment of slide quality, normalization, detection of differential gene expression and statistical analysis was carried out with ArrayPipe (version 1.6), a web-based, semi-automated software specifically designed for processing of microarray data (Hokamp K, et al. ArrayPipe: a flexible processing pipeline for microarray data. Nucleic Acids Res 2004; 32(Web Server issue):W457-9) (www.pathogenomics.ca/arraypipe). The following processing steps were applied: 1) flagging of markers, 2) subgrid-wise background correction, using the median of the lower 10% foreground intensity as an estimate for the background noise, 3) data-shifting, to rescue negative spots, 4) printTip LOESS normalization, 5) merging of technical replicates, 6) two-sided one-sample Student t-test on the log2-ratios within each treatment group, 7) averaging of biological replicates to yield overall fold-changes for each treatment group. Further, the gene expression data was overlaid on molecular interaction networks using Cytoscape (Shannon P, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 2003; 13:2498-504). Interactions networks were custom built from manually curated data and information contained within the Transpath pathway database (Krull M, et al. TRANSPATH: an integrated database on signal transduction and a tool for array analysis. Nucleic Acids Res 2003; 31:97-100). The false discovery rate of selecting differentially expressed genes from microarray analysis was estimated at 35%, based on Beta Uniform Mixture model (Pounds S, et al. Estimating the occurrence of false positives and false negatives in microarray studies by approximating and partitioning the empirical distribution of p-values. Bioinformatics 2003; 19:1236-42) and Q-Value model (Storey JD. A direct approach to false discovery rates. Journal of the Royal Statistical Society 2002; 64:479-498). This was consistent with the confirmation, using qPCR, at 4 different time points, of array results for 14 of 20 genes (70%) selected for follow-up.


Quantitative real-time PCR (qPCR)—Differential gene expression identified by microarray analysis was validated using quantitative real-time PCR (qPCR) using SuperScriptTM III Platinum® Two-Step qRT-PCR Kit with SYBR® Green (Invitrogen Life Technologies), as per the manufacturer's instructions, in the ABI PRISMS® 7000 sequence detection system (Applied Biosystems, Foster city, Calif., USA). Briefly, 1 μg of total RNA was reverse transcribed in a 20 μl reaction volume for 50 min at 42° C., the reaction was terminated by incubating for 5 min at 85° C. and then digested for 30 min at 37° C. with RNAse H. The PCR reaction was carried out in a 12.5 μl reaction volume containing 2.5 μl of 1/10 diluted cDNA template. A melting curve was performed to ensure that any product detected was specific to the desired amplicon. Fold changes were calculated after normalization to endogenous GAPDH and using the comparative Ct method (Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 2001; 29:No. 9 e45). The primers used for qRT-PCR are reported in Table 65.

TABLE 65Sequence of primers (human) used for qPCRGeneForward primer (5′-3′)Reverse Primer (5′-3′)CCL4CTTTTCTTACACCGCGAGGAAGCAGAGGCTGCTGGTCTCAT(SEQ ID NO: 67)(SEQ ID NO: 68)CCL20TGACTGCTGTCTTGGATACACAGATGATAGCATTGATGTCACAGCCT(SEQ ID NO: 69)(SEQ ID NO: 70)CXCL1GCCAGTGCTTGCAGACCCTGGCTATGACTTCGGTTTGGG(SEQ ID NO: 71)(SEQ ID NO: 72)IL-8GACCACACTGCGCCAACACCTTCTCCACAACCCTCTGCAC(SEQ ID NO: 73)(SEQ ID NO: 74)GAPDHGTCGCTGTTGAAGTCAGAGGGAAACTGTGGCGTGATGG(SEQ ID NO: 75)(SEQ ID NO: 76)IL-10GGTTGCCAAGCCTTGTCTGAAGGGAGTTCACATGCGCCT(SEQ ID NO: 77)(SEQ ID NO: 78)TNF-αTGGAGAAGGGTGACCGACTCTCCTCACAGGGCAATGATCC(SEQ ID NO: 79)(SEQ ID NO: 80)TNFAIP2CTACCAGCGCGCCTTTAATGTCCGGAAGGACAGGCAGTT(SEQ ID NO: 81)(SEQ ID NO: 82)TNFAIP3CTGCCCAGGAATGCTACAGATACCAGGGTCACCAAGGGTACAAA(SEQ ID NO: 83)(SEQ ID NO: 84)TNIP3TGAAAGAAAGGTAGCAGAGCTGAACCGCGTGCTGAGGAATCT(SEQ ID NO: 85)(SEQ ID NO: 86)BIRC3AAAGCGCCAACACGTTTGAAGGAACCCCAGCAGGAAAAG(SEQ ID NO: 87)(SEQ ID NO: 88)NF-κB1CTTAGGAGGGAGAGCCCACCTTGTTCAGGCCTTCCCAAAT(SEQ ID NO: 89)(SEQ ID NO: 90)RELATAGGAAAGGACTGCCGGGATCCGCTTCTTCACACACTGGA(SEQ ID NO: 91)(SEQ ID NO: 92)RELBTGGGCATTGACGCCTACAACTGGGTCCCTGAAGAACCATCAGGAAGTAGA(SEQ ID NO: 93)(SEQ ID NO: 94)NF-κBIAGGTGAAGGGAGACCTGGCTTGTGCCTCAGCAATTTCTGGC(SEQ ID NO: 95)(SEQ ID NO: 96)


Nuclear and Cytoplasmic Extracts—THP-1 cells (3×106) seeded into 60 mm2 petri dishes (VWR International, Mississauga, ON) were pre-treated with inhibitors for 30 min, and then stimulated with agonists or peptide for 30 min or 60 min. Cells were subsequently treated with Versene for 10 min at 37° C. in 5% CO2 (to detach adherent cells) then washed twice with ice-cold phosphate buffered saline. Cytoplasmic and nuclear extracts were isolated using NE-PER® Nuclear and Cytoplasmic Extraction Reagents Kit (Pierce Biotechnology, Rockford, Ill., USA) according to the manufacturer's instructions. The protein concentration of the extracts was quantified using a Bicinchoninic Acid (BCA) Protein Assay (Pierce Biotechnology) and the extracts were stored at −80° C. until fturther use.


Translocation of NFκB subunits—Equivalent nuclear extracts (5-10 μg) were resolved on a 7.5% SDS-polyacrylamide gel (SDS-PAGE) and transferred to polyvinylidene difluoride (PVDF) Immobilon-P membranes (Millipore Canada Ltd., Mississauga, ON). Equivalent protein loading was verified by staining PVDF membranes with Blot-Fast-Stain™ (Chemicon International) according to the manufacturer's instructions. Subsequently, the PVDF membranes were incubated with anti-p105/p50, anti-p65, anti-c-Rel, anti-Rel B or anti-p100/p52 antibodies at 1/1000 dilution in TBST (20 mM Tris pH 7.5, 150 mM NaCl, 0.1% Tween 20) containing 5% skimmed milk powder (TBST/milk) for 1 hr. Membranes were washed for 1 hour in TBST and then incubated with a 1/5000 dilution of HRP-conjugated goat anti-mouse or anti-rabbit Ab (in TBST/milk) for 30 min. The membranes were incubated for 30 to 60 min in TBST and developed with chemiluminescence peroxidase substrate (Sigma-Aldrich), according to manufacturer's instructions. Alternatively, equivalent nuclear extracts (2.5-10 ug) were analyzed for NFκB subunits p50 or p65 by StressXpress NFκB p50 or p65 ELISA kits (Stressgen Bioreagents, Victoria, BC, Canada) according to manufacturer's instructions. Luminescence was detected with SpectraFluor Plus Multifunction Microplate Reader (Tecan Systems Inc., SJ, USA).


Results


Low, physiological concentrations of SEQ ID NO: 1 suppress LPS-indauced secretion of the pro-inflarmnnatoiry cytokine TNFα. SEQ ID NO: 1 is found at mucosal surfaces at concentrations of around 2.5 to 5 μg/ml in adults and up to 20 μg/ml in infants (Schaller-Bals S, et al. Increased levels of antimicrobial peptides in tracheal aspirates of newborn infants during infection. Am J Respir Crit Care Med 2002; 165:992-5). Previous studies indicated that it has the ability to down-regulate pro-inflammatory cytokines in isolated monocytic cells (Bowdish D M, et al. Immunomodulatory activity of small host defense peptides. Antimicrob Agents Chemother 2005; 49:1727-32). To determine the lowest dose of SEQ ID NO: 1 that exhibited anti-endotoxin activity, THP-1 cells were stimulated with LPS (10 and 100 ng/ml) in the absence or presence of SEQ ID NO: 1 added simultaneously at concentrations ranging from 0.5 to 50 μg/ml for a period of 4 hours in complete RPMI cell culture media (i.e., which contains physiological salt concentrations). Tissue culture supernatants were assayed by ELISA for the presence of the pro-inflammatory cytokine TNFα (FIG. 6A). Very low concentrations (<1 μg/ml) of SEQ ID NO: 1 inhibited TNFα release from LPS-induced cells, demonstrating that physiological concentrations of SEQ ID NO: 1 exhibit anti-endotoxin activity. The anti-endotoxin effect of SEQ ID NO: 1 was more pronounced when the cells were stimulated with 10 ng/ml of LPS, a concentration at the lower level of concentrations used by investigators to mimic TLR signaling responses, but considerably higher than circulating endotoxin concentrations in septic patients (Opal S M, et al. Relationship between Plasma Levels of Lipopolysaccharide (LPS) and LPS-Binding Protein in Patients with Severe Sepsis and Septic Shock http://wwwjournals.uchicago.edu/JID/journal/issues/v180n5/990373/990373.text.html-fn1#fn1 J Infect Dis 1999; 180:1584-9). Under these conditions, 0.5 fg/ml of SEQ ID NO: 1 inhibited 50% of LPS-induced TNFα release. This inhibitory effect increased to ≧80% with a dose of 1 μg/ml of SEQ ID NO: 1, and TNFα was reduced to background levels with 2 μg/ml of SEQ ID NO: 1. In the presence of LPS at a higher concentration (100 ng/ml), 2 μg/ml of SEQ ID NO: 1 was required to inhibit 50% of TNFα released into the tissue culture supernatant. Higher concentrations (20 μg/ml) of SEQ ID NO: 1 caused ≧95% inhibition of TNFα release. These results indicated that physiological concentrations of SEQ ID NO: 1 exhibit an anti-endotoxin effect on LPS present at low and high concentrations. The anti-endotoxin effect of SEQ ID NO: 1 was similarly observed in PBMCs (FIG. 6B), for which SEQ ID NO: 1 (20 μg/ml) inhibited >91% of LPS (100 ng/ml) induced TNF-α. Subsequent mechanistic studies employed 100 ng/ml of LPS, at which concentrations more robust transcriptional up-regulation responses were observed, and 20 μg/ml of SEQ ID NO: 1, which was not cytotoxic to primary cells (Bowdish D, et al. The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. J Immunol 2004; 172:3758-65) or THP-1 cells as determined by LDH (lactose dehydrogenase) release and MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay (data not shown).


To gain further insight into the mode of inhibition exerted by SEQ ID NO: 1, TNFα production and release was monitored in the supernatants of LPS-stimulated THP-1 cells treated with the transcriptional inhibitor actinomycin D. Four μg/ml of actinomycin D was used since this concentration was required for inhibition, by more than 96% within 1 hour of treatment, of LPS-induced transcription of the genes for both the cytokine TNFα and the pro-inflammatory TNFα-inducible protein 2 (TNFAIP2) (monitored by qPCR, data not shown). Actinomycin D reduced the level of TNFα release by 97.6% (FIG. 6C), indicating that LPS largely induced de novo expression of TNFα as opposed to processing and release of intracellular pools of pro-form TNFα. Moreover, the use of monensin as an inhibitor of TNFα secretion led to accumulation of TNFα within cells after LPS stimulation for 60 min (FIG. 6D). However SEQ ID NO: 1 by itself did not similarly lead to the accumulation of TNFα inside cells, indicating that it also prevented TNFα expression at the protein level rather than blocking secretion.


The sustained presence of SEQ ID NO: 1 inhibits TNFα release. To determine the kinetics of the anti-endotoxin effect, the supernatant from THP-1 cells was monitored for TNFα after 1, 2, 4 and 24 hr of stimulation with LPS (100 ng/ml) in absence or presence of SEQ ID NO: 1 (20 μg/ml). When the peptide and LPS were added simultaneously, the release of TNFα was substantially inhibited (90 to 97%) by SEQ ID NO: 1 at all time points (FIG. 7A). When SEQ ID NO: 1 was added 30 min after LPS addition, TNFα secretion was reduced more than 50% at 2 and 4 hr post LPS treatment and by 80% after 24 hr (FIG. 7B) consistent with previous observations in mouse macrophages (Scott MG, et al. The human antimicrobial peptide SEQ ID NO: 1 is a multifunctional modulator of innate immune responses. J Imnmunol 2002; 169:3883-91). In contrast, when the cells were pre-treated with SEQ ID NO: 1 for 30 min, washed and stimulated with LPS, TNFα secretion was substantially (64%) reduced after 1 hr, but this declined to only 24 to 35% at subsequent time points (FIG. 7C). This indicated that a sustained presence of SEQ ID NO: 1 was required to exhibit a maximal anti-endotoxin effect.


SEQ ID NO: 1 suppresses TLR-induced cytokine secretion by PBMC. PBMC were treated with agonists of TLR2 (LTA, PAM3CSK4), TLR4 (LPS), TLR9 (CpG), and the inflammatory cytokines TNFα and IL-1β, to determine if SEQ ID NO: 1 could suppress cytokine secretion induced by inflammatory stimuli LPS and other agonists in primary cells. Cytokine production was analyzed by Luminex 100™ StarSystem using the human 5-Plex cytokine kit to monitor IL-1β, IL-6, IL-8 and TNFα in the culture supernatants. The cytokine profile of stimulated PBMC in the presence or absence of SEQ ID NO: 1 was monitored after 4 or 24 hours of treatment. The release of all 4 cytokines was significantly reduced by SEQ ID NO: 1 in both LPS- and LTA-stimulated cells after 4 hr of treatment, and this anti-inflammatory activity was sustained over 24 hr (FIG. 8). Effects on IL-8 production were more modest, as anticipated, since SEQ ID NO: 1 has the ability to induce IL-8 production (Scott MG, et al. The human antimicrobial peptide LL-37 is a multifunctional modulator of innate immune responses. J Immunol 2002; 169:3883-91). In addition, SEQ ID NO: 1 reduced IL-1β, IL-6, IL-8 and TNFα production by TLR2-agonist PAM3CSK4-stimulated PBMC after 4 or 24 hr of treatment, by approximately 30-50%, (Table 66). These data show that SEQ ID NO: 1 significantly reduced the production of pro-inflammatory cytokines resulting from activation of TLR2 or TLR4 (Table 66). SEQ ID NO: 1 also reduced, by ˜50%, IL-8 secretion by PBMC stimulated with the TLR9 agonist CpG for 24 hr (FIG. 8; Table 66).


In contrast, SEQ ID NO: 1 enhanced TNFα and IL-6 production by CpG-stimulated PBMC and IL-6, IL-8 and (modestly) TNFα by PBMC stimulated with IL-1β (FIG. 8; Table 66). Conversely, SEQ ID NO: 1 had no effect on TNFα induced cytokine production. These results indicate that the SEQ ID NO: 1 was anti-inflammatory in response to selected TLR ligands, and that it was likely modulating innate immune pathways rather than simply suppressing some step in the main TLR to NFκB pathway.


Table 66 lists percent inhibition or enhancement of agonist-induced cytokine production by SEQ ID NO: 1. PBMC were incubated alone or with TLR agonists (LPS, LTA, CpG) or inflammatory cytokines (TNFα, IL-1β) for 4 or 24 hr in the presence or absence of SEQ ID NO: 1. The concentration of IL-1β, IL-6, IL-8 and TNFα released in the tissue culture supernatant is reported. The percent inhibition of IL-1β, IL-6, IL-8 and TNFα in the presence of SEQ ID NO: 1±the standard deviation of 3 biological repeats is reported, as well as the fold enhancement of cytokine production in the presence of SEQ ID NO: 1±the standard deviation of 3 biological repeats.

AgonistCells OnlyTNF-αIL-1βLPSAve pg/mlAve pg/mlAve pg/mlAve pg/ml−SEQ+SEQFold Inc.or−SEQ+SEQFold Inc.−SEQ+SEQFold Inc.−SEQ+SEQFold Inc.IDID% Inh.IDIDor % Inh.IDIDor % Inh.IDIDor % Inh.NO: 1NO: 1(Ave ± SD)NO: 1NO: 1(Ave ± SD)NO: 1NO: 1(Ave ± SD)NO: 1NO: 1(Ave ± SD)Release by4 hrIL-1β<974711.0 ± 0.1N/A34<9>81.9 ± 17.7IL-6<7<739531.2 ± 0.4435<7>98.4 ± 0.8IL-815322.1 ± 0.154901.9 ± 0.71243332.4 ± 1.273884  89.1 ± 7.7TNF-α<16N/A979N/A83073  96.2 ± 2.2Release by24 hrIL-1β<994951.0 ± 0.2N/A51214  99.1 ± 1.3IL-6<79131.7 ± 0.664548158.1 ± 1.47734170  97.9 ± 1.3IL-83753210.3 ± 4.0441053201.4 ± 0.6303484522.9 ± 0.976203332  74.7 ± 15.8TNF-α<16N/A2045123.6 ± 12.62334303  78.9 ± 18.0AgonistCpGLTAPAM3Fold Inc.Ave pg/mlAveor % Inh.−SEQFold Inc. orpg/ml(Ave ± SD)ID+SEQ ID% Inh.−SEQ ID+SEQ IDFold Inc. or %−SEQ+SEQ IDFold Inc. or %NO: 1NO: 1(Ave ± SD)NO: 1NO:1Inh. (Ave ± SD)ID NO: 1NO: 1Inh. (Ave ± SD)Release by4 hrIL-1β53<9>87.0 ± 14.534<9>81.9 ± 17.7<9IL-6139124  98.3 ± 0.8435<7>98.4 ± 0.8<717.0IL-81366273  79.8 ± 11.873884  89.1 ± 7.720341.7 ± 0.6TNF-α183666  96.3 ± 0.683073  96.2 ± 2.228343.5 ± 2.6Release by24 hrIL-1β969<9>99.4 ± 0.651214  99.1 ± 1.3<96.6 ± 1.7IL-614887318  97.9 ± 1.57734170  97.9 ± 1.366417IL-871082928  58.8 ± 23.176203332  74.7 ± 15.833917448.6 ± 1.0 TNF-α404039  99.2 ± 0.62334303  78.9 ± 18.02817117.6 ± 20.5


LPS-induced gene expression profile is altered by SEQ ID NO: 1. Human 21K oligo-based DNA microarrays were probed to elucidate the impact of SEQ ID NO: 1 on LPS stimulation of gene responses in human monocytic cells. Transcriptional responses were analyzed following 1, 2, 4 and 24 hr of stimulation to provide a temporal profile of gene expression in monocytes equivalent to the early, intermediate and late stages of innate immune responses. Microarray analyses were performed in duplicate from three independent biological replicates. Statistically significant, differentially expressed genes were defined as those with a fold change of at least 1.5 with a Student's t-test p-value≦0.05 (MIAME compliant data was deposited to ArrayExpress). The number of differentially expressed genes was greatest at the 2 and 4 hr time points. Over the monitored time period, 561 and 410 genes were differentially regulated in the presence of LPS, without or with SEQ ID NO: 1 respectively. Of the 561 genes that were differentially expressed in LPS-stimulated cells, only 39 (˜7%) were identified as being up-regulated in cells stimulated with LPS in the presence of SEQ ID NO: 1 (Table 67). At least 163 genes that were upregulated in cells stimulated with LPS (i.e., proinflaninatory genes) were suppressed in the presence of SEQ ID NO: 1 (Table 68). This indicates that SEQ ID NO: 1 effectively suppressed the induction of a large subset of LPS-responsive genes, but maintained a modest subset of genes that function in promoting some aspects of inflammation or anti-inflammatory response.

TABLE 67List of 39 genes differentially expressed upon stimulation by LPS andremaining up-regulated in the presence of SEQ ID NO: 1, as detected by microarrayanalysis at one or more time points.LPS_1 hrLPS_2 hrLPS_4 hrLPS_24 hrFoldFoldFoldFoldGene Namechangep-valuechangep-valuechangep-valuechangep-valueZNF834.210.011.590.59−1.420.73−1.200.92NFKBIA1.710.012.220.351.880.111.530.05Q9P1881.690.021.130.241.660.093.300.24INVS1.690.02−1.360.601.510.731.440.87DIAPH11.770.02−1.490.871.810.13−1.150.58IER31.580.032.260.101.990.032.920.12Q9H6401.620.041.430.44−1.450.53−1.930.36GBP21.320.052.100.012.380.021.040.34NANS1.130.051.650.041.620.071.810.00Q86XN7;2.670.068.010.047.450.03−1.020.20Q9H9M1TNFAIP32.470.073.350.053.710.041.330.23Q96MJ8;1.740.084.010.011.900.581.650.05Q9BSE2Q9H7532.290.083.910.022.550.771.020.75NTNG13.750.08−1.460.271.050.411.520.02INHBE1.580.091.840.05−1.070.641.070.73BCL61.760.121.670.031.730.041.050.25CXCL12.540.124.260.051.980.111.300.39EHD11.800.133.420.053.170.021.880.08RELB1.160.142.160.052.800.021.420.22HRK1.820.151.580.233.150.502.720.05CCL42.030.152.430.011.710.091.200.15SESN21.260.172.470.052.660.03−1.330.57NAB11.220.171.670.052.460.061.170.31EBI31.180.195.590.061.780.12−1.060.40DDX211.260.231.510.062.740.15−1.080.35XBP11.760.231.800.051.320.051.390.08SULRP1; ARS1.560.252.100.171.330.231.800.05HDAC102.190.311.350.191.600.061.130.25MEP1A−1.230.391.080.72−1.160.592.470.02RAP2C1.340.431.700.032.610.041.370.09GYS1−1.300.47−1.010.542.170.032.260.51RARRES31.290.48−2.190.571.010.661.770.05PPY1.190.491.710.611.581.004.280.02NFKB11.160.751.720.011.890.03−1.120.97MTL4_HUMAN1.100.811.520.042.220.23−1.070.88Q9H040−1.620.82−1.020.721.580.011.710.43Q9NUP61.510.991.310.281.250.126.860.06LPS + SEQLPS + SEQLPS + SEQLPS + SEQID NO: 1ID NO: 1ID NO: 1ID NO: 11 hr2 hr4 hr24 hrFoldFoldFoldFoldGene Namechangep-valuechangep-valuechangep-valuechangep-valueZNF832.020.031.080.651.170.41−1.370.38NFKBIA1.940.032.360.011.500.231.300.02Q9P1881.580.041.870.322.140.022.050.15INVS1.550.02−2.950.081.770.961.440.08DIAPH12.070.01−1.520.962.770.041.780.13IER31.510.042.150.021.550.431.350.36Q9H6401.770.021.480.37−1.990.21−1.970.10GBP21.720.08−1.290.361.510.061.330.33NANS1.020.761.010.51−1.410.271.700.04Q86XN7;1.670.203.710.041.080.411.780.14Q9H9M1TNFAIP32.500.143.450.022.340.041.200.67Q96MJ8;1.630.031.860.261.690.892.620.00Q9BSE2Q9H7531.150.212.320.001.120.771.310.24NTNG11.550.111.290.271.090.533.390.06INHBE−1.010.672.570.01−1.060.56−1.240.39BCL61.020.221.950.011.200.481.200.81CXCL11.930.124.560.032.080.631.090.49EHD11.640.133.480.001.550.151.730.07RELB−1.020.252.580.002.000.931.110.20HRK3.460.082.011.002.280.872.090.05CCL41.360.191.880.051.800.051.140.86SESN2−1.050.881.300.161.620.011.120.45NAB1−1.090.472.420.001.410.03−1.200.66EBI3−1.250.541.960.021.890.472.440.26DDX211.210.371.550.001.600.011.310.05XBP11.120.091.580.00−1.020.321.020.68SULRP1; ARS2.620.461.200.301.390.511.850.02HDAC101.220.241.320.861.970.011.320.32MEP1A−1.850.112.050.101.220.751.890.06RAP2C1.270.291.540.031.310.501.080.22GYS1−1.150.75−1.180.171.960.05−1.020.46RARRES3−1.130.461.150.701.240.132.620.05PPY−4.350.482.500.261.130.695.650.04NFKB11.200.781.650.051.450.931.020.44MTL4_HUMAN−1.260.871.520.011.180.081.030.41Q9H040−1.190.89−1.260.521.510.00−1.530.22Q9NUP61.310.591.290.90−1.270.641.900.01









TABLE 68










Genes that are upregulated by the Toll-like receptor 4 ligand LPS and


downregulated by LL-37.














LPS +





LPS
LL37
LL37




fold
fold
fold


Gene Name
Gene Description
change
change
change














LC2A6
Facilitative glucose transporter; binds cytochalasin B with low affinity
7.04
1.13
1.41


SLC4A5
HCO3-transporter; Na+/HCO3-co-transporter
6.80
1.52
4.72


MCL1
Apoptosis regulator Bcl-2 protein, BH
6.31
1.73
1.72


Q86XN7; Q9H9M1
Aldehyde dehydrogenase; Proline-rich extensin; Proline-rich region
6.00
1.41
2.29


Q86UU3; Q8NAA1
Proline-rich extensin; Proline-rich region
5.41
−1.08
1.16


C15orf2
low complexity
5.24
−2.56
−1.29


TNFRSF5
Receptor for TNFSF5/CD40L
5.24
−1.30
1.82


FACL6
Activation of long-chain fatty acids for both synthesis of cellular lipids, and degradation
5.09
1.50
2.61



via beta-oxidation.


Q8IW99; Q96AU7
Thymic Stromal Lymphopoietin Isoform 2.
4.92
−1.12
−1.20


PRB4
Salivary proline-rich protein II-1
4.9
−1.02
−1.29


Q9NWP0
low complexity
4.89
−1.20
−1.06


Q8NF24; Q8TEE5
β-Ig-H3/Fasciclin domain; Proline-rich extension
4.60
1.45
1.06


PDE4DIP
Similar to Rat Myomegalin.
4.56
1.27
−1.42


NUDT4
Nudix hydrolase
4.55
−1.33
−1.39


DUSP2
Regulates mitogenic signal transduction by dephosphorylating both Thr and Tyr
4.42
1.35
1.46



residues on MAP kinases ERK1 and ERK2


LMAN2
Intracellular lectin in the early secretory pathway; transport and sorting of high
4.38
−1.41
−1.37



mannose-type glycoproteins


RELB
Stimulates promoter activity in the presence of p49- and p50-NFκB. Neither associates
4.30
1.96
1.23



with DNA nor with p65-NFκB


SNF1LK
Probable serine/threonine-protein kinase SNF1LK
4.27
1.25
1.93


TNFα
Cytokine that binds to TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR.
4.25
1.14
2.64


GHRHR
G protein-coupled receptor for growth hormone GRF.
4.11
−3.22
1.01


TNFSF6
Cytokine that binds to TNFRSF6/FAS, a receptor that transduces the apoptotic signal
3.79
1.32
1.69



into cells.


ENSG00000181873
Glycine cleavage T protein (aminomethyl transferase)
3.78
−1.18
1.96


IRAK2
Required for IL1R-induced NFκB activation. Proximal mediators of IL-1 signaling
3.71
1.41
1.46


CKB
Reversibly catalyzes the transfer of phosphate between ATP and various phosphogens
3.60
1.39
1.57



(e.g. creatine phosphate).


CASR
Senses changes in the extracellular concentration of calcium ions.
3.51
1.01
−1.47


KRTAP4-10
Keratin, high sulfur B2 protein; von Willebrand factor, type C
3.45
1.69
−3.16


ARHGEF3
DH domain; Pleckstrin-like
3.43
1.01
1.10


CYP3A4; CYP3A7
P450 Cytochrome.
3.43
−4.24
−1.00


GPR27
Orphan receptor. Possible candidate for amine-like G-protein coupled receptor
3.41
1.25
−1.83


PAX8
Transcription factor for the thyroid-specific expression of the genes.
3.37
−1.95
−5.99


GAP43
Associated with nerve growth. Major component of the motile & growth cones
3.36
1.87
−1.81


Q96M75; Q9H568
Actin/actin-like
3.31
−2.73
1.50


AGTRL1
Receptor for apelin coupled to G proteins that inhibit adenylate cyclase activity.
3.24
2.00
1.24



Alternative co-receptor with CD4 for HIV-1 infection.


C1orf22
Putative α-mannosidase C1orf22
3.21
1.17
1.11


EHD1
EH-domain containing protein 1; Testilin; hPAST1
3.20
1.58
1.6


ADRA1B
G protein-coupled α-adrenergic receptor
3.17
1.62
−1.60


SSTR2
G protein-coupled receptor for somatostatins-14 and -28.
3.17
1.09
1.27


SYNE1
Involved in the maintenance of nuclear organization and structural integrity. Connects
3.16
1.37
−1.30



nuclei to the cytoskeleton.


ENSG00000139977
Bipartite nuclear localization signal; GCN5-related N-acetyltransferase
3.15
−1.94
−1.20


PTPRK
Regulator of processes involving cell contact and adhesion such as growth control,
3.13
1.33
1.19



tumor invasion, and metastasis.


O15059; Q9NZ16
Guanine-nucleotide dissociation stimulator CDC25; Pleckstrin-like
3.13
1.28
3.43


N4BP3; KIAA0341
Nedd4-binding protein 3; N4BP3
3.11
−1.28
1.60


Q8IVT2
coiled-coil; low complexity
3.10
1.32
−1.73


Q9NV39
low complexity
3.08
−1.39
−1.72


HIP1R; HIP12;
Component of clathrin-coated pits and vesicles, may link the endocytic machinery to
3.06
−1.22
1.21


KIAA0655
actin cytoskeleton


IL-6
Cytokine with a wide variety of biological functions
3.04
1.11
1.46


TNFAIP2
May play a role as a mediator of inflammation and angiogenesis; Probably function in
2.97
1.54
1.0



nuclear protein import as nuclear transport receptor.


RCV1
Seems to be implicated in the pathway from retinal rod guanylate cyclase to rhodopsin.
2.95
−1.38.
−1.69


FBLN2
Its binding to fibronectin and some other ligands is calcium dependent
2.95
1.14
−1.04


TWIST2
Inhibits transcriptional activation by MYOD1, MYOG, MEF2A and MEF2C. Represses
2.92
1.80
2.05



expression of proinflammatory cytokines such as TNFα and IL1β.


PARD6B
Adapter protein involved in asymmetrical cell division and polarization processes and
2.88
−3.02
1.46



formation of epithelial tight junctions.


DCK
Required for the phosphorylation of several deoxyribonucleosides.
2.84
1.23
1.65


TULP4
Tubby-like protein 4; Tubby superfamily protein
2.83
−2.18
1.07


KLK10
Has a tumor-suppressor role for NES1 in breast and prostate cancer
2.81
1.40
1.25


SPAP1
Immunoglobulin-like
2.80
1.23
2.35


IBRDC2
Zn-finger, RING; Zn-finger, cysteine-rich C6HC
2.79
−1.64
1.03


JAM2
May play a role in the processes of lymphocyte homing to secondary lymphoid organs
2.77
−2.6
−1.44


NRG2
Direct ligand for ERBB3 and ERBB4 tyrosine kinase receptors. May also promote the
2.74
−1.44
2.31



heterodimerization with the EGF receptor


CBARA1
Bipartite nuclear localization signal; Calcium-binding EF-hand
2.74
1.5
1.74


DLG2
Interacts with the cytoplasmic tail of NMDA receptor subunits as well as potassium
2.66
1.55
−1.0



channels


PRKCBP1
Protein kinase C binding protein 1
2.66
−3.68
−1.42


MGLL
Alpha/beta hydrolase; Alpha/beta hydrolase fold; Esterase/lipase/thioesterase, active
2.65
1.56
1.07



site; Lipase


Q9BYE1
Chymotrypsin serine protease, family S1; Low density lipoprotein-receptor, class A;
2.60
−2.52
−3.84


MARCKS
MARCKS is the most prominent cellular substrate for protein kinase C. Binds
2.60
1.33
1.13



calmodulin, actin, and synapsin and is an F-actin cross-linking protein


Q96N98
Amidase
2.60
1.25
1.07


Q8NBY1; Q96AF2;
Bipartite nuclear localization signal; Protein kinase; Tyrosine protein kinase
2.60
1.28
1.30


Q9BS16
Soxlz/Sox6-binding protein SolT.
2.58
−2.57
1.82


PPP2CA
Protein phosphatase PP2A can modulate the activity of MAP-2 kinase and other
2.58
−1.47
1.19



kinases.


RAB38
May be involved in melanosomal transport and docking. Involved in the proper sorting
2.54
−1.778
1.62



of TYRP1


VCAM1
Important in cell-cell recognition. VCAM1/VLA4 interaction may play a role in
2.53
1.46
2.21



immune responses and in leukocyte emigration to inflammation sites


TTTY8
Transcript Y 8 protein
2.52
1.22
−1.13


HTR2A
One of the several different serotonin G protein-coupled receptors
2.51
−1.20
−1.35


SERPINB10
May play a role in the regulation of protease activities during hematopoiesis
2.51
1.51
−5.00


O75121; Q9BVE1
Immunoglobulin-like
2.51
−2.15
−1.07


ZCCHC2
Phox-like; Zn-finger, CCHC type
2.50
−1.04
1.60


CXCL2
Chemokine produced by activated monocytes & neutrophils and expressed at
2.50
1.38
1.42



inflammation sites


GADD45B
Involved in the regulation of growth & apoptosis. Mediates activation of
2.48
1.29
1.17



MTK1/MEKK4 MAPKKK


KARS
Lysy1-tRNA synthetase LysRS
2.43
1.29
−2.94


SCG2
Secretogranin II; a neuroendocrine secretory granule protein, biologically active peptide
2.42
−1.83
1.45



precursor


SLC17A2
May be involved in actively transporting phosphate into cells via Na(+) cotransport
2.41
1.03
1.08


FLT4
Receptor for VEGFC. Has a tyrosine-protein kinase activity
2.41
1.41
2.48


Q9NXT0
KRAB box; Zn-finger, C2H2 type
2.38
1.01
−1.22


Q96L19
L-lactate dehydrogenase;
2.38
1.00
1.12


BICD1

Drosophila Bicaudal D Homolog 1

2.34
−1.66
−4.36


HCK
May also contribute to neutrophil migration and may regulate the neutrophil
2.32
1.72
1.11



degranulation


Q8N9T8; Q9H978
Krr1
2.31
−1.26
−2.64


PPP1R1A
Inhibitor of protein-phosphatase 1.
2.31
−3.64
1.33


PAX7
Probable transcription factor. May have a role in myogenesis
2.31
−1.01
1.52


EBI3
Cytokine receptor
2.29
1.69
2.00


THRA
Nuclear hormone receptor. High affinity receptor for triiodothyronine
2.29
−3.93
−1.63


SLC16A10
Solute carrier family 16 (Monocarboxylate transporters), member 10
2.25
−1.72
6.63


INPP5E
Endonuclease/exonuclease/phosphatase family; Prenyl group binding site (CAAX box)
2.25
1.16
2.82


Q9H967
Bipartite nuclear localization signal; G-protein beta WD-40 repeat
2.23
1.50
3.75


NFKB1
NFκB1 p105 and p50 subunits involved in immune response and acute phase reactions.
2.21
1.36
1.09


MKL1
Antiapoptotic transcriptional factor that acts as a cofactor of serum response factor
2.21
1.24
−1.08



(SRF).


SS18L2
SS18-like protein 2; SYT homolog-2
2.17
1.16
1.09


TNFRSF9
Receptor for TNFSF14/4-1BBL. Possibly active during T cell activation
2.16
1.02
−1.37


TNFAIP6
Possibly involved in cell-cell and cell-matrix interactions during inflammation &
2.16
1.55
−1.17



tumorgenesis


Q9Y2K2
Protein kinase; Serine/Threonine protein kinase; Tyrosine protein kinase
2.14
1.16
1.12


ING5
Zn-finger-like, PHD finger
2.11
1.77
1.12


IL1A
Pro-inflammatory cytokine.
2.11
1.35
−2.22


TMH
unknown
2.10
−1.15
1.38


HDAC4
Histone deacetylase acts on lysine residues on the N-terminus of core histones.
2.10
−1.44
−1.02


KPTN
Kaptin actin-binding protein.
2.10
1.41
2.98


SEC61G
Necessary for protein translocation in the endoplasmic reticulum
2.07
−1.14
4.02


Q9Y484
G-protein beta WD-40 repeat
2.07
1.08
−2.49


FRAS1
von Willebrand factor, type C Cytochrome c heme-binding site; Signal peptidase;
2.05
−3.27
2.13


IER5
Immediate early response 5.
2.01
−1.06
1.37


Q8N137; Q8NCB8
LysT-interacting protein Lip8.
2.01
−1.16
2.01


Q96HQ0; Q9H5P0
ATP/GTP-binding site motif A (P-loop); KRAB box; Zn-finger, C2H2 subtype;
2.00
−1.31
1.94


TXNRD1
Thioredoxin reductase, cytoplasmic precursor; TR; TR1
1.99
1.17
1.06


CAV2
Caveolin-2; May act as a scaffolding protein within caveolar membranes.
1.98
−1.17
−1.48


SCARB1
CD36 antigen
1.97
−1.16
2.25


MAP3K5
Phosphorylates and activates two different subgroups of MAP kinase kinases.
1.96
1.16
1.375


PDHX
Required for anchoring dihydrolipoamide dehydrogenase (E3) to pyruvate
1.96
1.32
1.23



dehydrogenase


TCEB3
SIII, or elongin, is a general transcription elongation factor.
1.95
1.07
2.51


C21orf55
May have a role in protein folding or as a chaperone
1.95
1.07
2.03


MPHOSPH10
Component of U3 nucleolar small nuclear ribonucleoprotein. Processing preribosomal
1.94
1.19
1.22



RNA


PDE8A
Phosphodiesterase plays a role in signal transduction by regulating the intracellular
1.93
−1.33
1.17



concentration of cyclic nucleotides.


TFR2
Transferrin receptor 2. Cellular iron uptake o
1.92
−1.57
1.60


FARP1
Band 4.1 domain; DH domain; Pleckstrin-like
1.92
1.26
10.39


SERPINA1
Inhibitor of serine proteases. Primary target is elastase. Moderate affinity for plasmin,
1.92
1.30
1.23



thrombin


MYO15A
Myosins-15A; Unconventional myosins serve in intracellular movements.
1.91
1.32
−1.59


RABGGTA
Catalyzes the transfer of a geranyl-geranyl moiety from geranyl-geranyl pyrophosphate
1.89
1.27
−1.22



to both cysteines in certain Rab proteins.


KCNMB4
Calcium-activated BK potassium channel, beta subunit
1.89
1.12
1.56


Q9BR02
Bipartite nuclear localization signal; Ribosomal protein L23, N-terminal domain
1.89
−1.08
1.54


APOB
Apolipoprotein B; Recognition signal for the cellular binding and internalization of
1.88
1.39
−1.48



LDL.


MYC
Binds DNA both in a non-specific manner and activates transcription of growth-related
1.87
1.23
1.1



genes


FARP2
Band 4.1 domain; DH domain; Pleckstrin-like
1.85
1.32
1.12


TFAP2BL1
Transcription factor AP-2
1.84
1.22
2.04


Q86U90; Q9H5F8
SUA5/yciO/yrdC, N-terminal
1.82
1.07
−1.01


USH1C
May be involved in protein-protein interaction
1.81
−1.29
1.22


SOX2
Transcription factor SOX-2
1.78
1.32
−1.19


Q9NVC3
Amino acid/polyamine transporter, family II
1.78
−1.57
2.57


NEIL2
Formamidopyrimidine-DNA glycolase
1.76
−1.21
1.91


TNIP1
Interacts with TNFAIP3 and inhibits TNF-induced NFκB-dependent gene expression
1.75
1.41
1.09


ADRA1D
This alpha-adrenergic receptor mediates its effect through the influx of extracellular
1.72
−1.96
−1.0792



calcium


PCDHB9
Potential calcium-dependent cell-adhesion protein.
1.72
−2.70
1.96


Q12987
Bipartite nuclear localization signal
1.71
−1.06
1.18


TNFRSF6
Receptor for TNFSF6/FASL.
1.71
1.49
1.75


C20orf72
Protein C20orf72
1.70
1.14
1.67


DNAJA3
Modulates apoptotic signal transduction or effector structures within the mitochondrial
1.69
−1.20
−1.26



matrix.


MAB2IL1
Guanylate kinase; Mab-21 protein
1.67
−3.06
−1.43


BIRC2
Apoptotic suppressor. Interacts with TRAF1 and TRAF2.
1.67
1.34
1.12


MYST1
MOZ/SAS-like protein
1.66
1.32
3.50


CNN3
Thin filament-associated protein
1.66
1.00
1.12


CXCL3
Chemokine: May play a role in inflammation.
1.65
−2.13
−1.215


CD80; CSRP2;
Involved in the costimulatory signal essential for T lymphocytes activation.
1.65
−1.07
1.13


RAD51L1


ADARB1; TNFSF8
Cytokine that binds to TNFRSF8/CD30. Induces proliferation of T cells;
1.64
−1.04
−3.34


Q8IW74
unknown
1.62
1.09
−1.02


UXS1
NAD-dependent epimerase/dehydratase
1.62
1.11
−1.04


ENSG00000182364;
Phosphatidylinositol 3- and 4-kinase
1.61
−1.46
−1.19


TNFRSF7
Receptor for TNFSF7/CD27L. May play a role in survival of activated T-cells.
1.60
1.29
−1.25


MYBL2
Transcription factor involved in the regulation of cell survival, proliferation, and
1.60
−1.07
−1.22



differentiation.


RAB33A
Ras-related protein Rab-33A; Small GTP-binding protein S10
1.60
−1.30
1.15


ATIC
Bifunctional purine biosynthesis protein PURH;
1.59
−1.36
−1.166


CAMK1
Phosphorylates synapsin I
1.59
1.26
1.53


CCNT1
Regulatory subunit of the cyclin-dependent kinase pair (CDK9/cyclin T) complex
1.58
1.17
1.97


KCNE4
β subunit of voltage-gated potassium channel complex of pore-forming alpha subunits.
1.57
−1.20
1.41


BOK
Apoptosis regulator Bcl-2 protein,
1.56
−1.21
1.12


NF2
Probably acts as a membrane stabilizing protein
1.56
1.27
1.36


PDP2; KIAA1348
Catalyzes the dephosphorylation/reactivation of the α-subunit of pyruvate
1.51
−2.13
−1.12



dehydrogenase E1 component









Given that LPS has been known to induce inflammatory responses via the TLR4 to NFκB pathway (Chow JC, et al. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem 1999; 274:10689-92) and the product of certain differentially expressed genes in the microarray analysis were associated with this pathway, we analyzed in more detail the NFκB-regulated genes and the TLR4 pathway. This pathway was first mapped by integrating protein:protein interaction, signal transduction and regulatory data from the literature into Cytoscape (www.cytoscape.org), an open-source bioinformatics software platform for visualizing molecular interaction networks and integrating these interactions with other data. The microarray expression data was then overlaid onto this signal transduction protein network by colour coding the individual nodes (equivalent to specific genes/proteins) according to the extent of regulation (ranging from red to green, where the intensity of colour demonstrated the extent of up- to down regulation respectively). This then provided a graphic illustration of the genes with altered expression in response to LPS in the absence or presence of SEQ ID NO: 1 at each of the time points (FIG. 9A), and indicated that LPS generally up-regulated genes encoding elements of the TLR4→NFκB pathway, with a peak response at 2-4 hours, and that SEQ ID NO: 1 generally dampened this up-regulation.


To investigate further whether a defined portion of the LPS-responsive genes were likely co-regulated by NFκB, LPS-responsive, differentially-expressed genes with similar temporal expression profiles were clustered using the K-means procedure, a non-hierarchical algorithm, with an affinity threshold of 85% (FIG. 9B). Each cluster thus represented a set of potential co-regulated genes (based on their similar expression profiles over time). Based on this method, the LPS-induced genes were divided into 15 clusters. Three of these clusters, containing a total of 123 genes with peak expression at 2 hr, 4 hr or both, contained 21 genes that are known from the literature to be NFκB-regulated (FIG. 9B). On the other hand, the temporal expression patterns of the 410 genes induced by LPS in the presence of SEQ ID NO: 1 fell into 8 clusters, one of which contained 11 of the 12 differentially expressed NFκB gene targets; six of these NFκB target genes were also included in the subset of LPS-stimulated genes and demonstrated modestly to substantially decreased expression in the presence of SEQ ID NO: 1. Many p50/p65 target genes (Tian B, et al. Identification of direct genomic targets downstream of the NF-kappa B transcription factor mediating TNF signaling. J Biol Chem 2005) were found in the clusters containing the NFκB genes. Thus SEQ ID NO: 1 clearly resulted in the suppression of LPS-stimulation of a substantial number of known NFκB target genes, and clustering data indicated that many other genes that might be NFκB regulated were similarly suppressed. However the data also suggested that the effect observed was selective in that some known NFκB regulated genes were still apparently differentially expressed in the presence of the combination of LPS and SEQ ID NO: 1. To confirm these observations, genes with significant differential expression in response to LPS, and that were differentially affected (remained up-regulated or abrogated) by the presence of the peptide, were selected for validation by quantitative real-time PCR.


SEQ ID NO: 1 selectively modulates the transcription of specific LPS-induced inflammatory genes. Using qPCR, the expression profiles were validated for 14 of 20 selected genes differentially expressed according to the microarray analysis (FIG. 10). Several known “pro-inflammatory” genes were up-regulated after 2 and 4 hr of treatment with LPS, and this expression level invariably decreased after 24 hr of stimulation. Further, the expression of several LPS-induced genes was confirmed to be altered by the presence of SEQ ID NO: 1. Even though the peptide had a dampening effect on selected LPS-induced expression of inflammatory genes, not all genes up-regulated by LPS were suppressed by the presence of SEQ ID NO: 1, indicating that the effect of SEQ ID NO: 1 on LPS-induced inflammation was selective (FIG. 10). The expression of pro-inflammatory genes such as NFκB1 (p 105/p50) and TNFAIP2 were substantially reduced (90-97%) in LPS-stimulated cells in the presence of SEQ ID NO: 1 at all time points. Also, LPS-induced transcription of TNFα was reduced in the presence of SEQ ID NO: 1 by 87% after 1 hr and around 80% at 2 and 4 hr, but at 24 hr only 58% reduction was observed. Similarly, LPS-induced transcription of IL10 was reduced by more than 90% after 1 and 2 hr in presence of SEQ ID NO: 1, and this effect decreased to 77% after 4 hr. In contrast, the expression of chemoattractants such as IL-8, CCL4, and CXCL1, was slightly reduced by SEQ ID NO: 1 in LPS-stimulated cells but not completely eliminated. Likewise, the expressions of certain anti-inflammatory genes, that are negative regulators of the TLR4 to NFκB pathway were only slightly reduced in the presence of SEQ ID NO: 1. These genes included TNFαIP3 (TNFα-inducible Protein 3) and its interacting partner TNIP3 (TNFαIP3-interacting protein 3), as well as the NFκB-inhibitor, NFκBIA. LPS-induced transcription of NFκB subunit NFκB1 (p105/p50), but not RelB, was completely abrogated by SEQ ID NO: 1, whereas RelA (p65) did not show significant differential expression in response to LPS or SEQ ID NO: 1.


From the temporal transcriptional profiling of LPS-induced genes, it was concluded that SEQ ID NO: 1 did not substantially affect the LPS-induced expression of selected genes that are required for cell recruitment and movement (chemokines) or negative regulators of NFκB. In contrast, SEQ ID NO: 1 neutralized the expression of genes coding for inflammatory cytokines, NFκB1 (p105/p50) and TNFα-induced pro-inflarnmatory genes such as TNFAIP2.


SEQ ID NO: 1 significantly inhibits LPS-induced translocation of the NFκB subunits p50 and p65. The above data indicated that although LL-37 reduced TNFα secretion by more than 95% at all time points, it had a lesser effect (58-87%) in reducing TNFα transcription. To study this in more detail we investigated the key transcription factor NFκB. TLR activation results in nuclear translocation of NFκB, the key transcription factor required for expression of many innate immunity and inflammatory genes (Bonizzi G, et al. The two NF-B activation pathways and their role in innate and adaptive immunity. Trends Immunol 2004; 25:280-8; Li Z W, et al. Genetic dissection of antigen receptor induced-NF-kappaB activation. Mol Immunol 2004; 41:701-14). Although NFκB has a number of subunits with different primary transcriptional regulatory functions, the p50/p65 NFκB heterodimer is most commonly implicated in the regulation of immunity genes. Nevertheless, transcriptionally active NFκB heterodimers other than p50/p65 have important functions as it has been shown that they can influence gene responses to bacterial molecules as well as susceptibility to a variety of infections (Tato CM, et al. Host-Pathogen interactions: Subversion and utilization of the NF-κB pathway during infection. Infect Immunity 2002; 70:3311-7; Mason N, et al. Cutting edge: identification of c-Rel-dependent and -independent pathways of IL-12 production during infectious and inflammatory stimuli. J Immunol 2002;168:25904). To determine if SEQ ID NO: 1 suppressed LPS-induced changes in gene expression by affecting NFκB translocation into the nucleus, the nuclear localization of five NFκB subunits was assessed by Western blots. All monitored subunits of NF-κB (p105/50, p65, c-Rel, Rel B and p100/52) were detected in the nuclear extracts of THP-1 cells (FIG. 11A). The nuclear localization of p50, p65, c-Rel and Rel B, and to a lesser extent p100/52, was increased in THP-1 cells stimulated with LPS for 30 and 60 min (by 60 mins, LPS had induced a 3.5 fold increase in nuclear p5O, a 4.5 fold increase in p65, a 1.7 fold increase in RELB and c-REL, and a 1.2 fold increase in p100/52 as assessed by densitometry). The LPS-induced translocation of p50, p65 and Rel B was clearly suppressed in the presence of SEQ ID NO: 1 as there was around a 35-70% decrease in subunit translocation after 60 min (FIG. 11A), while p100/52 and c-Rel did not appear to be affected.


To more accurately quantify the translocation of p50 or p65, the nuclear extracts were analyzed by ELISA-based immunoassays specific for these subunits (FIG. 11B). SEQ ID NO: 1 suppressed, by slightly more than 50%, LPS-induced p50 and p65 translocation at 30 and 60 min (54±4% and 56±4% inhibition of p50 at 30 and 60 min respectively and 57±8% and 54±3% inhibition of p65 at 30 and 60 min respectively). As a control, it was demonstrated that polymyxin B, a known inhibitor of LPS-LBP (LPS-binding protein) engagement, more substantially inhibited the translocation of NFRB subunits p50 and p65 (82±5% and 80±90% respectively at 60 min; data not shown), demonstrating that TLR4 to NFκB activation can be blocked significantly by agents acting at the cell surface. Although SEQ ID NO: 1 has been reported to activate signal transduction pathways including MAPK in human monocytes and lung epithelial cells (Bowdish D, et al. The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. J Immunol 2004; 172:3758-65), SEQ ID NO: 1 did not promote translocation of NF-κB subunits in human THP-1 cells. Together, these data demonstrate that SEQ ID NO: 1 can moderately alter the LPS-induced translocation of NFκB subunits, thereby providing one mechanism by which SEQ ID NO: 1 suppressed pro-inflammatory cytokine production.


To evaluate the anti-endotoxic activity of SEQ ID NO: 1, two different concentrations of LPS, 10 ng/ml and 100 ng/ml respectively, were used to stimulate human monocytic cells in the presence or absence of this host defense peptide, in an attempt to reflect concentrations of endotoxin ranging from the presumably low concentrations secreted by the normal flora (homeostatic conditions) and early in infection, to those observed in septic infections. To date there has been considerable controversy concerning the role of SEQ ID NO: 1 in human infections, particularly at physiological concentrations. Direct antimicrobial action will certainly occur at low salt concentrations but in the presence of more physiological concentrations of Na+(130 mM) and Mg2+/Ca2+(1-2 mM) found in tissues and in tissue culture medium (as employed here), SEQ ID NO: 1 has weak or no direct antimicrobial action at the peptide concentrations (1-5 μg/ml) apparently present at mucosal surfaces (Bowdish DM, et al. Impact of SEQ ID NO: 1 on anti-infective immunity. J Leukoc Biol 2005; 77:451-9). Nevertheless there is clear evidence of an anti-infective role (Scott MG, et al. The human antimicrobial peptide LL-37 is a multifunctional modulator of innate immune responses. J Immunol 2002; 169:3883-91; Bowdish DM, et al. Impact of LL-37 on anti-infective immunity. J Leukoc Biol 2005; 77:451-9; Kirikae T, et al. Protective effects of human 18-kilodalton cationic antimicrobial protein (CAP-18)-derived peptide against murine endotoxemia. Infect Immun 1998; 66:1861-8; Fukumoto K, et al. Effect of antibacterial cathelicidin peptide CAP 18/LL-37 on sepsis in neonatal rats. Pediatr Surg Int 2005; 21:20-4; Ciornei CD, et al. Antimicrobial and chemoattractant activity, Lipopolysaccharide neutralization, cytotoxicity, and inhibition by serum of analogs of human cathelicidin LL-37. Antimicrob Agents Chemother 2005; 49:2845-50), which could be explained if SEQ ID NO: 1 has a role in modulating innate immunity. Consistent with this concept, at physiological concentrations SEQ ID NO: 1 is able to mediate chemotaxis (Agerberth B, et al. The human antimicrobial and chemotactic peptides LL-37 and alpha-defensins are expressed by specific lymphocyte and monocyte populations. Blood 2000; 96:3086-93; Yang D, et al. Participation of mammalian defensins and cathelicidins in anti-microbial immunity: receptors and activities of human defensins and cathelicidin (LL-37). J Leukoc Biol 2001; 69:691-7; Niyonsaba F, et al. A cathelicidin family of human antibacterial peptide LL-37 induces mast cell chemotaxis. Immunology 2002;106:20-6), MAP kinase phosphorylation (Scott MG, et al. The human antimicrobial peptide LL-37 is a multiftuctional modulator of innate immune responses. J Immunol 2002; 169:3883-91; Tjabringa G S, et al. The antimicrobial peptide LL-37 activates innate immunity at the airway epithelial surface by transactivation of the epidermal growth factor receptor. J Immunol 2003; 171:6690-6; Bowdish D, et al. The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. J Immunol 2004;. 172:3758-65; Lau YE, et al. Interaction and cellular localization of the human host defense peptide LL-37 with lung epithelial cells. Infect Immun 2005; 73:583-91), Ca2+mobilization (Niyonsaba F, et al. Evaluation of the effects of peptide antibiotics human beta-defensins-1/-2 and LL-37 on histamine release and prostaglandin D(2) production from mast cells. Eur J Immunol; 2001; 31:1066-75) and IL-8 release in GM-CSF treated monocytes (Bowdish D M, et al. Impact of LL-37 on anti-infective immunity. J Leukoc Biol 2005; 77:451-9), and as shown herein, anti-endotoxic activity.


The sole human cathelicidin peptide, SEQ ID NO: 1, has been shown to protect animals against endotoxemia/sepsis. Low, physiological concentrations of SEQ ID NO: 1 (≦1 μg/ml) are able to modulate inflammatory responses by inhibiting the release of the pro-inflammatory cytokine TNFα in LPS-stimulated human monocytic cells. Microarray studies established a temporal transcriptional profile, and identified differentially expressed genes in LPS-stimulated monocytes in the presence or absence of SEQ ID NO: 1. SEQ ID NO: 1 significantly inhibited the expression of specific pro-inflammatory genes upregulated by NFκB in the presence of LPS, including NFκB1 (p105/p50) and TNFα-induced protein 2 (TNFAIP2). In contrast, SEQ ID NO: 1 did not significantly inhibit LPS-induced genes that antagonize inflammation, such as TNFα-induced protein 3 (TNFAIP3) and the NFκB inhibitor, NFκBIA, or certain chemokine genes that are classically considered pro-inflammatory. Nuclear translocation, in LPS-treated cells, of the NFκB subunits p50 and p65 was reduced >50% in the presence of SEQ ID NO: 1, demonstrating that the peptide altered gene expression in part by acting directly on the TLR to NFκB pathway. SEQ ID NO: 1 almost completely prevented the release of TNFα and other cytokines by human peripheral blood mononuclear cells (PBMC) following stimulation with LPS and other TLR2/4 and TLR9 agonists, but not with cytokines TNFα or IL1β. Biochemical and inhibitor studies were consistent with a model whereby SEQ ID NO: 1 modulated the inflammatory response to LPS/endotoxin and other agonists of TLRs by a complex mechanism involving multiple points of intervention.


The data presented herein conclusively demonstrates that endotoxin-induced inflammatory gene responses and cytokine secretion in monocytes were suppressed by low, physiological concentrations of SEQ ID NO: 1, implicating SEQ ID NO: 1 in the regulation and control of pro-inflammatory responses associated with pathogenic assault and, by extension, with homeostatic levels of TLR agonists secreted by commensals. The data further demonstrates that SEQ ID NO: 1 can suppress LPS-induced NFκB translocation, and exert an anti-inflammatory effect that is not restricted to endotoxin-induced inflammation. In the human THP-1 monocytic cell line as well as in human PBMC, SEQ ID NO: 1 suppressed pro-inflammatory cytokine production induced by LPS as well as other agonists of TLR2 (LTA, PAM3CSK4) and in part TLR9 (CpG), but selectively enhanced responses to the pro-inflammatory cytokines IL 1β and TNFα. To gain mechanistic insight, transcriptional responses were profiled using microarrays and real time PCR over the course of 1 to 24 hr to study the effects of SEQ ID NO: 1 on LPS-stimulated monocytes. While the transcription of LPS-induced pro-inflammatory cytokines peaked at 2-4 hr and waned by 24 hr, a single, low dose of SEQ ID NO: 1 suppressed pro-inflammatory cytokine secretion by 1 hr, and this effect was sustained for 24 hr.


Overall, the data provides evidence that SEQ ID NO: 1 can manipulate both pre- and post-transcriptional events to modulate the TLR-induced inflammatory response in monocytes. A model consistent with the data in this manuscript is outlined in FIG. 12.


LPS-induced activation of NFκB is mediated by TLR4, a receptor containing TIR domain. It is known that receptors with TIR domains are potent activators of NFκB, as well as several other transcription factors such as AP-1, NF-IL6 and IRF3/7 (Takeda K, et al. Toll receptors and pathogen resistance. Cell Microbiol 2003;5:143-53). Mice deficient in TLR4 or MD2 are hyposensitive to LPS, moreover expression of some NFκB target genes is defective without MD2 (Poltorak A, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 1998; 282:2085-8; Hoshino K, et al. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J Immunol 1999;162:3749-52; Nagai Y, et al. Essential role of MD-2 in LPS responsiveness and TLR4 distribution. Nat Immunol 2002;3:667-72). NFκB is known to play a central role in pathogenesis resulting in sepsis (Brown M A, et al. NF-kappaB action in sepsis: the innate immune system and the heart. Front Biosci 2004; 9:1201-17; Xiao C, et al. NF-kappaB, an evolutionarily conserved mediator of immune and inflammatory responses. Adv Exp Med Biol 2005; 560:41-5) as well as innate immunity to infections (Alcamo E, et al. Targeted mutation of TNF receptor I rescues the RelA-deficient mouse and reveals a critical role for NF-kappa B in leukocyte recruitment. J Immunol 2001; 167:1592-600; Senftleben U, et al. IKKbeta is essential for protecting T cells from TNFalpha-induced apoptosis. Immunity 2001;14:217-30). NFκB transcription factor is a dimeric complex of various subunits that belong to the Rel family; p105/50 (NFκB1), p100/52 (NFκB2), p65 (RelA), RelB, and c-Rel. NFκB proteins share a 300-amino acid Rel homology domain (RHD) that contains a nuclear localization sequence (NLS) and is involved in dimerization, sequence-specific DNA binding and interaction with the inhibitory IkB proteins (Ghosh S, et al. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol 1998; 16:225-26). The NFκB proteins form numerous homo- and hetero-dimers that are associated with specific biological responses that stem from their ability to regulate target gene transcription differentially, e.g., p50/p52 dimers function as repressors, whereas Rel A or c-Rel dimers are transcriptional activators. In contrast, RelB does not form homodimers, but instead forms stable heterodimers with either p50 or p52 to exhibit a greater regulatory flexibility, and can be either an activator (Ryseck R P, et al. RelB, a new Rel family transcription activator that can interact with p50-NF-kappa B. Mol Cell Biol 1992; 12:674-84) or a repressor (Ruben S M, et al. I-Rel: a novel rel-related protein that inhibits NF-kappa B transcriptional activity. Genes Dev 1992; 6:745-60). Many inflammatory stimuli trigger signal transduction pathways that result in nuclear localization of NFκB and subsequent transcription of inflammatory and immunity genes encoding for cytokines, chemokines, acute phase reactants, and cell adhesion molecules. The NFκB heterodimer comprising of p50 and p65 subunits has been strongly implicated in transcriptional events triggered by the activation of pro-inflammatory cytokine receptors or TLRs (Ghosh S, et al. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol 1998; 16:225-260; Wang T, et al. NF-kappa B and SpI elements are necessary for maximal transcription of toll-like receptor 2 induced by Mycobacterium avium. J Immunol 2001; 167:6924-32). The activation and nuclear translocation of NFκB p50/p65 heterodimer is associated with increased transcription of genes encoding chemokines, cytokines, adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and endothelial-leukocyte adhesion molecule 1 (ELAM), as well as enzymes that produce secondary inflammatory mediators and inhibitors of apoptosis (Ghosh S, et al. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol 1998; 16:225-260). These molecules are important components of the innate immune responses to invading pathogens and are required for migration of inflammatory mediators and phagocytic cells to tissues where NFκB has been activated in response to infection or injury (Pande V, et al. NF-kappaB in human disease: current inhibitors and prospects for de novo structure based design of inhibitors. Curr Med Chem 2005; 12:357-74).


The present invention provides evidence that the host defense peptide, SEQ ID NO: 1, can partially (˜50%) reduce LPS-induced p50/p65 translocation to the nucleus, indicating that this is one mechanism whereby SEQ ID NO: 1 suppressed LPS-induced gene transcription and exerted an anti-endotoxin effect. However if SEQ ID NO: 1 were merely blocking the binding of LPS to the TLR4 receptor through inhibiting its interaction with LBP and/or the LPS receptor complex (Scott M G, et al. Cutting edge: cationic antimicrobial peptides block the binding of lipopolysaccharide (LPS) to LPS binding protein. J Immunol 2000; 164, 549-53), it would be expected that NFκB translocation, and all NFκB-dependent transcriptional events would be inhibited to the same extent as TNFα release, that is>95%; however, this was not observed here. Instead, the effects of SEQ ID NO: 1 on NFκB subunit translocation were selective and relatively modest, and effects on LPS-stimulated transcription of NFκB-regulated genes ranged from very high, e.g., >95% for TNFαIP2 and p105/p50, to moderate (˜80%) for TNFα itself, through to almost no inhibition for other NFκB-regulated genes like TNFAI3. Similarly SEQ ID NO: 1 can protect against sepsis in animal models when administered shortly after endotoxin (Bowdish D, et al. The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. J Immunol 2004; 172:3758-65). In unpublished mouse model experiments (K. Lee, M. G. Scott and R. E. W. Hancock), it was demonstrated that 200 μg of SEQ ID NO: 1 could protect against an 80% lethal dose (400 μg) of E. coli LPS administered peritoneally. Under such circumstances, the LPS would be in 5-fold molar excess and it seems unlikely that in this situation LPS neutralization alone could explain the protection exhibited by SEQ ID NO: 1.


The data presented herein indicates that the host defense peptide SEQ ID NO: 1 can selectively regulate genes that modulate inflammatory responses by suppressing NFκB translocation leading to dysregulation (modulation) of TLR-triggered transcriptional responses. SEQ ID NO: 1 caused inhibition of LPS-triggered pro-inflammatory gene TNFAIP2, but did not neutralize the LPS-induced expression of some of the known negative regulators of NFκB such as TNFAIP3, TNIP3 and NFκBIA (IκBα). Conversely, the transcription of known LPS-induced genes that are regulated by p50/p65 (FIG. 9B) were also inhibited >90% in the presence of SEQ ID NO: 1. However, although NFκB transcription factor activity is influenced by changes in nuclear concentration and subunit composition, the observed ˜50% inhibition of p50/p65 translocation in LPS-induced cells by SEQ ID NO: 1 seems unlikely to completely account for the observed 80% reduction in TNFα gene transcription at 2-4 hr or the >95% reduction in TNFα: protein production and release. Rather, this nearly complete inhibition of pro-inflammatory cytokine release, without an equivalent abrogation of gene transcription, implies that mechanisms other than inhibition of NFκB are also required for SEQ ID NO: 1 to regulate TLR-induced inflammation. Such anomalies demonstrate that SEQ ID NO: 1 influences post-transcriptional events to modulate the inflammatory response. It is therefore shown that SEQ ID NO: 1 affects components of protein translation, maturation or secretion directly and/or indirectly via SEQ ID NO: 1—activated effectors or SEQ ID NO: 1—induced gene transcription (FIG. 12). It is known that SEQ ID NO: 1 can activate components of the MAPK pathway, in particular, p38 (which can influence post-transcriptional events) and ERK, and can promote the activity of the transcription factor, Elk-1 (Bowdish D, et al. The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. J Immunol 2004; 172:3758-65). The putative receptors for SEQ ID NO: 1, including FPRL-1, P2X7, and EGRFR, do not appear to be responsible for SEQ ID NO: 1 induced activation of the MAPK pathway in monocytes (Bowdish D, et al. The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. J Immunol 2004; 172:3758-65). In Drosophila, the LPS or PGN mediated up-regulation of expression of NFκB dependent genes is reported to be suppressed by a MAPK-regulated transcription factor, AP-1 (Kim T, et al. Downregulation of lipopolysaccharide response in drosophila by negative crosstalk between the AP1 and NF-κB signaling modules Nature Immunology 6, 211-218 (2005)). SEQ ID NO: 1 also demonstrates synergy with inflammatory stimuli such as GM-CSF (Bowdish D M, et al. Impact of LL-37 on anti-infective immunity. J Leukoc Biol 2005; 77:451-9; Devine D A, et al. Cationic peptides: distribution and mechanisms of resistance. Curr Pharm Des 2002; 8:703-14) and IL1 P (FIG. 8; Table 66) that likely reflect activation of co-operative signal transduction pathways or transcription of genes whose products contribute to a stabilized, enhanced or prolonged response. Thus, SEQ ID NO: I probably works alone or synergistically with other effector molecules of innate immunity, potentially via the MAPK pathway, to modulate TLR activation and enhance host defense mechanisms.


Accordingly, the data demonstrates that SEQ ID NO: 1 selectively suppresses the pro-inflammatory response in monocytes, particularly the TLR-induced secretion of pro-inflammatory cytokines. The ability of SEQ ID NO: 1 to dampen pro-inflammatory (septic) responses would be valuable for maintaining hormeostasis in the face of natural shedding of microflora-associated TLR agonist molecules, as well as limiting the induction of systemic inflammatory syndrome/septic shock in response to moderate pathogen challenge. The anti-inflammatory effects of SEQ ID NO: 1 were observed at physiologically relevant concentrations of the peptide, and small changes in peptide concentration led to substantial impact on the cellular response to bacterial components such as LPS. SEQ ID NO: 1 thus appears to manifest multiple, complex mechanisms of action, including direct and indirect inhibition of TLR activation and transcription. The improved understanding of the mechanism(s) utilized by SEQ ID NO: 1 to selectively modulate inflammation, and thereby balance the TLR response to commensal or pathogenic bacteria indicates that endogenous cationic host defense peptides are important players in limiting over-active inflammation.


EXAMPLE 14
Analysis of Transcriptional Responses Elicited by Synthetic Host Defense Peptides

Endogenous host defense peptides are widely distributed in nature, are essentially amphipathic in nature, 12-50 amino acids in length with a net positive charge of +2 to +7 and are about 50% hydrophobic (Hancock, et al. 1999. Peptide antibiotics. Antimicrob Agents Chemother. 43:1317-1323). As shown above, certain natural host defense peptides expressed in mammalian epithelial and a variety of myeloid cells have been demonstrated to be selectively anti-inflammatory, and are able to maintain the expression of genes that are key players in innate immunity such as certain chemokines, as well as others that antagonize inflammation. In these activities, the peptides are able to act synergistically with other immune mediators such as GM-CSF (FIG. 4) and IL1β. The human peptide LL-37 (SEQ ID NO: 1) induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes. (FIGS. 3 and 4). Some of these peptides also have the potential to exert overall anti-inflammatory responses, but others have specific therapeutic deficits including the ability to induce apoptosis in certain cells and the stimulation of histamine release through degranulation of mast cells. However, the specificity and efficacy of host defense peptides that can selectively modulate innate immune responses can be improved by deriving synthetic peptides based on the motifs of natural host defense peptides using both random as well as rational design (Hilpert, et al. 2005. High-throughput generation of small antibacterial peptides with improved activity. Nat Biotechnol. 23:1008-1012). Therefore, this study investigated global transcriptional-responses elicited by the synthetic host defense peptide KSRIVPAIPVSLL (SEQ ID NO: 7) in order to unravel the signaling pathways activated by the peptides themselves. The objective was to evaluate the effects of host defense peptides on innate immunity, thereby permitting elucidation of the mechanisms of action as well as biomarkers for peptide action.


The overall effects of synthetic peptide SEQ ID NO: 7 on CD 14+monocytes isolated from human peripheral blood mononuclear cells (PBMC) were studied by using a functional genomics and bioinformatics approach. The global transcriptional responses elicited by the peptide, and its effect on bacterial endotoxin-stimulated cells were elucidated in human monocytic cells. Gene profiling technology using DNA microarrays, followed by other transcriptional analysis, e.g., Real-time PCR, and functional analyses applied by the investigators have provided new insight into the molecular events underlying the mechanism elicited by synthetic cationic host defense peptides.


Materials and Methods


The venous blood from healthy volunteers was collected in Vacutainere collection tubes containing sodium heparin as an anticoagulant (Becton Dickinson, Mississauga, ON). White blood cells were isolated from-the buffy coat, and the PBMC was seeded maintained in physiologically relevant RPMI-1640 media (Gibco®, Invitrogen™ Life technologies, Burlington, ON), supplemented with 10% (v/v) heat inactivated fetal bovine serum (FBS), 2 mM L-glutamine and 1 mM sodium pyruvate (all from Invitrogen Life Technologies) at 37° C. in a humidified 5% (v/v) C02 incubator. Human monocytic cells, THP-1 (ATCC TIB-202) were grown in suspension in RPMI-1640 complete media as described above. They were treated with 0.3 μg/ml phorbol 12-myristate 13-acetate (PMA; Sigma-Aldrich Canada, Oakville ON) for 24 hr, inducing plastic-adherent cells that were further rested in complete RPMI-1640 medium for an additional 24 hr prior to stimulations with various treatments.


The cells were stimulated with synthetic host defense peptide SEQ ID NO: 7 (200 μg/ml), as well as purified bacterial LPS/endotoxin (2 ng/ml) in the presence and absence of synthetic peptide SEQ ID NO: 7 for 4 hours. The cells were treated with the synthetic peptide 45 mins prior to stimulation with bacterial LPS. Highly purified LPS free of proteins and lipids was obtained from P. aeruginosa strain H103 using the Darveau-Hancock method as previously described (Darveau, et al. 1983. Procedure for isolation of bacterial lipopolysaccharides from both smooth and rough Pseudomonas aeruginosa, Salmonella enterica ssp., and Typhimurium strains. J Bacteriol. 155: 831-838). The synthetic peptide SEQ IID NO: 7 was synthesized using F-moc chemistry.


Following incubation of the cells, the tissue culture supernatants were centrifuged at 1000×g for 5 min, then at 10,000×g for 2 min to obtain cell-free samples, aliquoted and stored at −20° C. prior to assay for various cytokines. Protein secretion were detected with a capture ELISA (eBioscience and BioSource International Inc., CA, USA respectively) using the tissue culture supernatants. In addition, CD14+ monocytes were isolated from the stimulated PBMC after incubation using magnetic Dynal® bead-based separation technology (Invitrogen™). RNA was isolated from the purified monocytes using RNeasy Mini kit, treated with RNase-Free DNase (Qiagen Inc., Canada) and eluted in RNase-free water (Ambion Inc., Austin, Tex., USA) as per the manufacturer's instructions. RNA concentration, integrity and purity were assessed by Agilent 2100 Bioanalyzer using RNA 6000 Nano kits (Agilent Technologies, USA). RNA was (reverse) transcribed with incorporation of amino-allyl-UTP (aa-UTP) using the MessageAmpII™ amplification kit, column purified and eluted in nuclease-free water, and labeled with mono-functional dyes, Cyanine-3 and Cyanine-5 (Amersham Biosciences), according to manufacturer's instructions. Yield and fluorophore incorporation was measured using Lambda 35 UV/VIS fluorimeter (PerkinElmer Life and Analytical Sciences, Inc., USA).


Microarray slides were printed with the human genome 21K Array-Ready Oligo Set™ (Qiagen Inc., USA) at The Jack Bell Research Center (Vancouver, BC, Canada). The slides were pre-hybridized and scanned as described (Mookherjee, et al. 2006. Modulation of the TLR-Mediated Inflammatory Response by the Endogenous Human Host Defense Peptide LL-37. J Immunol. 176: 2455-2464). Assessment of slide quality, normalization, detection of differential gene expression and statistical analysis was carried out with ArrayPipe (version 1.6), a web-based, semi-automated software specifically designed for processing of microarray data (www.pathogenomics.ca/arraypipe). Differentially expressed and statistically significant genes were selected from the miocroarray analysis as genes that induced an absolute fold change of at least 1.5 with a p-student value of ≦0.06.


Differential gene expression identified by microarray analysis was validated using quantitative real-time PCR (qPCR) using SuperScript™ III Platinum® Two-Step qRT-PCR Kit with SYBR® Green (Invitrogen Life Technologies), as described (Mookherjee, et al.). Fold changes were calculated after normalization to endogenous human GAPDH and using the comparative Ct method (Pfaffl, M. W. 2001. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29: No. 9 e45).


Results and Discussion


Gene profiling using human 21K human DNA microarrays revealed that there were 566 genes differentially expressed in human monocytes in presence of SEQ ID NO: 7 (Table 69). Of these genes those that were significantly up-regulated by the peptide included G-coupled protein receptors, other transmembrane receptors and co-activators, genes associated with the plasma membrane including integrin, adhesion molecules such as ICAM, NCAM, genes that regulate various tyrosine-protein kinases, critical transcription factors that mediate key pathways in immune responses such as MAPK, JAK-STAT and NFκB. The peptide also up-regulated the expression of various chemotatic factors that play a central role in attracting immune cells to the site of infection and/or trauma. Other significant genes induced by the peptide were those encoding transport proteins including those involved in metal transport, several zinc finger proteins fimctioning as transcription factors, and several genes that are attributed to anti-viral activity.


The genes that were significantly upregulated by the peptide included:

  • (a) G-coupled protein receptors that initiate signaling from extracellular ligands—representative genes include, but are not limited to, GPR55, GPR6, GPR30, GPCR42, CASR, EDG2;
  • (b) Chemokines and Interleukins that attract immune cells—representative genes include, but are not limited to, MCP-1, MCP-3, IL-8, IL-17C, as well as receptors for chemokines, e.g., CCR7;
  • (c) Transcription factors that mediate selective gene expression—representative genes include, but are not limited to, JAK1, STAT1, ELF1, Q9Y4C1, ETV4, POU1F1, Zinc finger proteins e.g., ZNF254, ZNF292, ZNF78L1, Homeobox transcription factor e.g., HOXD3, DLX5;
  • (d) Tyrosine-protein kinase and their receptors—representative genes include, but are not limited to MAP2K6, NTRK3, PLCG1, EFNA2, NCK1;
  • (e) Adhesion molecules that mediate cell attachment and interaction—representative genes include, but are not limited to, ICAM, NCAM, as well as cell adhesion receptors e.g., PTPRF;
  • (f) Genes involved in actin polymerization and cytoskeletal remodelling involved in cell movement and differentiation—representative genes include, but are not limited to, Integrin-α, EPHA4, ARHGAP6, and DST.
  • (g) Regulators of transcription facotors—representative genes include, but are not limited to, TRIP4, GMEB2, GSK3B, ARNT, BACH1, ARID3A, HIPK2, POLR2D, TGIF, SSBP3, FYB;
  • (h) Transmembrane receptors and adapters of signaling pathways including, but not limited to WNT5B, receptor for WNT proteins FZD10, TIRAP (adapter for TLR4 pathway), REPS1; and


(i) Genes involved in antiviral activity—representative genes include, but are not limited to Interferons e.g., IFNA2, STAT1 that activates gamma interferon, transcriptional activator MNDA, Interferons that exhibit anti-viral activities-representative genes include, but are not limited to IFNA2.

TABLE 69Gene profiling using human 21K human DNA microarrays revealing that therewere 566 genes differentially expressed in human monocytes in presence of SEQ ID NO: 7Fold ChangeGene NameGene DescriptionSEQ ID NO: 7P-valueO43300Leucine-rich repeat80.00.05TGM4Associated with mammalian reproductive71.90.04process. Catalyzes the cross-linking ofproteins and the conjugation of polyaminesto specific proteins in the seminal tractTMOD4Blocks the elongation/depolymerization of68.00.04the actin filaments at the pointed end.Q86Y93ATP/GTP-binding site motif A (P-loop);64.30.05CSL zinc finger; Heat shock protein DnaJRBP1Intracellular transport of retinol63.30.04Q9C098Protein kinase; Serine/Threonine protein59.00.05kinaseQ8NI35ATP/GTP-binding site motif A (P-loop);57.20.05PDZ/DHR/GLGF domainQ8WUC6Bipartite nuclear localization signal; Class I46.00.05peptide chain release factor domainARNTRequired for activity of the Ah (dioxin)45.20.04receptor. This protein is required for theligand-binding subunit to translocate fromthe cytosol to the nucleus after ligandbinding. This complex then initiatestranscription of genes involved in theactivation of PAH procarcinogensTNCSAM (substrate-adhesion molecule) that45.10.05appears to inhibit cell migration. May playa role in supporting the growth of epithelialtumors. Ligand for integrins α-8/β-1, α-9/β-1, α-V/β-3 and α-V/β-6POU1F1Transcription factor involved in the44.50.04specification of the lactotrope,somatotrope, and thyrotrope phenotypes inthe developing anterior pituitary. Activatesgrowth hormone and prolactin genes.FEZ2Involved in axonal outgrowth and40.20.02fasciculationSMURF2E3 ubiquitin-protein ligase which accepts38.60.06ubiquitin from an E2 ubiquitin-conjugatingenzyme in the form of a thioester and thendirectly transfers the ubiquitin to targetedsubstrates, SMAD1, SMAD2 and SMAD7KCNK6Exhibits outward rectification in physiological36.80.04K(+) gradient & mild inwardrectification in symmetrical K(+)conditionsSMFSMF protein36.80.04CNTN5Fibronectin, type III; Immunoglobulin-like33.60.05O43348Argininosuccinate synthase33.20.03WBSCR18Williams-Beuren syndrome chromosome33.00.05region 18 proteinQ9Y4T9low complexity31.70.05ZNF595Maybe transcriptional repressor. Candidate31.50.05gene for Wolf-Hirschhorn (4p-) syndromeCD226Immunoglobulin-like31.00.04BNIP1Implicated in the suppression of cell death.29.70.05Interacts with the BCL-2 and adenovirusE1B 19 kDa proteinsIDUAAlpha-L-iduronidase precursor29.20.06Q8N6Q6unknown27.10.05IL17CStimulates the release of TNFα and IL-1β26.50.05from the monocytic cell line THP-1monocytic cell line THP-1RRM2Catalyzes the biosynthesis of deoxyribo-24.60.04nucleotides from the correspondingribonucleotides for DNA synthesisNR2F2Regulation of the apolipoprotein A-I gene24.20.03transcription. Binds to DNA site ADLX5Homeobox protein DLX-523.40.04CHRNA1After binding acetylcholine, the AChR23.30.03responds by opening of an ion-conductingchannel across the plasma membraneSLC35A4Nucleotide-sugar transporter23.20.05TRPM8Ion transport protein22.60.04RNF130Basic helix-loop-helix dimerization domain22.50.05bHLH; Bipartite nuclear localizationsignal; Protease-associated PA; Zn-finger,RINGDKC1Required for ribosome biogenesis and21.80.05telomere maintenance. Probable catalyticsubunit of H/ACA small nucleolarribonucleoprotein complex, whichcatalyzes pseudouridylation of rRNA.LRMPLymphoid-restricted membrane protein.21.50.06HOOK2Probable cytoskeletal linker protein, which21.40.03may be involved in tethering membranebound organelles to the cytoskeletonDHX8Facilitates nuclear export of spliced mRNA21.20.04by releasing the RNA from the spliceosomeQ86WW9ATP/GTP-binding site motif A (P-loop);20.80.06Lipoxygenase, LH2 domainQ9P278low complexity20.20.04STATHSalivary protein that stabilizes saliva20.20.06supersaturated with Ca2+ salts; modulateshydroxyapatite crystal formation in teethQ96LW2Blue (type 1) copper domain; Protein20.10.05kinase; Serine/Threonine protein kinaseSBNO1Helicase, C-terminal; RNA-binding region19.70.05RNP-1 (RNA recognition motif)IDH3GIsocitrate dehydrogenase [NAD]subunit19.40.02gamma, mitochondrial precursor; Isocitricdehydrogenase; NAD+-specific ICDHQ9H6R7coiled-coil; low complexity19.00.04PLCG1phospholipase C-γ is a major substrate for18.40.05heparin-binding growth factor 1-activatedtyrosine kinaseKIAA1529coiled-coil; low complexity;18.10.04transmembraneQ8NHU6Bipartite nuclear localization signal;17.70.04Maternal tudor proteinRCL1Plays a role in 40S-ribosomal-subunit17.40.01biogenesis in the early pre-rRNAprocessing steps at sites A0, A1 and A2:required for proper maturation of 18S RNAQ96HJ9unknown16.90.05FHL3Four and a half LIM domains protein 3;15.20.04FHL-3; Skeletal muscle LIM-protein 2;SLIM 2Q8N8U9Trypsin inhibitor-like, cysteine-rich TIL14.80.03region; von Willebrand factor, type C & DO60384Zn-finger, C2H2 type14.80.05ENSG00000105849RNA polymerase Rpa43 subunit14.80.05MCART1Mitochondrial carrier triple repeat 114.60.05BCL2Suppresses apoptosis in a variety of cell14.10.04systems. Regulates cell deathEFNA2Ephrin-A2 precursor; EPH-related receptor14.10.04tyrosine kinase ligand 6; LERK-6;Q9H697limkain beta 2.13.90.04PTGER3Receptor for prostaglandin E2 (PGE2)13.30.05CKMT1Reversibly catalyzes the transfer of13.10.05phosphate between ATP and variousphosphogens (e.g. creatine phosphate).TUBB4QTubulin; the major constituent of13.10.06microtubules.Q7Z483Zn-finger, RING12.80.04DUSP14Involved in the inactivation of MAP12.70.05kinases. Dephosphorylates ERK, JNK andp38 MAP-kinasesCYLC2Acc: Q14093]; Cylicin II (Multiple-band12.50.03polypeptide II). [Source: SWISSPROTOR5P2Putative odorant receptor. Could also be12.20.04involved in taste perceptionEPB41L4BBand 4.1-like protein 4B; EHM2 protein;11.80.02FERM-containing protein CG1IL17BStimulates the release of tumor necrosis11.60.03factor alpha and IL-1 beta from themonocytic cell line THP-1VPS4AAAA ATPase, central region; ATP/GTP-11.50.04binding site motif A (P-loop); MITC6orf74Metal-dependent phosphohydrolase, HD11.30.06regionSYNE1Involved in the maintenance of nuclear11.30.06organization and structural integrity.Probable anchoring protein which thetersthe nucleus to the cytoskeleton.ZNF322BZn-finger, C2H2 type10.90.04SACM1LSynaptojanin, N-terminal10.90.04PRO0461Protein PRO046110.70.01ALOX5Arachidonate 5-lipoxygenase10.60.04Q7RTU0Basic helix-loop-helix dimerization domain10.40.05bHLHTRPM3Calcium channel mediating constitutive10.10.03calcium ion entry.POMT2Transfers mannosyl residues to the9.90.06hydroxyl group of serine or threonineresidues.PCSK5Likely widespread endoprotease activity9.90.04within the constitutive and regulatedsecretory pathway. Cleaves RX(K/R)RMAP3K1Component of protein kinase signal9.80.05transduction cascade. Activates the ERKand JNK kinase pathwaysas well as CHUKand IKBKB, the central protein kinases ofthe NFκ-B pathwayBLZF1Basic leucine zipper nuclear factor 1.9.80.06Q96D46Cytochrome c heme-binding site; KRAB9.60.03box; NMD3 familyQ9NW81Leucine-rich repeat9.40.05Q9BUJ0Alpha/beta hydrolase;9.20.03Esterase/lipase/thioesterase, active siteQ9H5P1Zn-finger, C-x8-C-x5-C-x3-H type9.10.05UBE2NThe UBE2V2/UBE2N heterodimer9.10.04catalyzes the synthesis of non-canonicalpoly-ubiquitin chains that are linkedthrough Lys-63. This type of poly-ubiquitination does not lead to proteindegradation by the proteasome. Mediatestranscriptional activation of target genes.Plays a role in the control of progressthrough the cell cycle and differentiation.Plays a role in the error-free DNA repairpathway and contributes to the survival ofcells after DNA damageO75872rab3 GTPase-activating protein, non-8.80.03catalytic subunit.ANKRD5Ankyrin repeat domain protein 58.60.05MAN2B2Epididymis-specific alpha-mannosidase8.30.04pre-cursor; Mannosidase alpha class 2Bmember 2RPS740S ribosomal protein S7; 40S ribosomal8.30.05protein S7; S8PDE7BMay be involved in the control of cAMP-8.30.03mediated neural activity and cAMPmetabolism in the brainBACH2Component of the dystrophin-glycoprotein8.20.04complex (DGC), a complex that spans themuscle plasma membrane and forms a linkbetween the F-actin cytoskeleton and theextracellular matrix. Probably involved inthe control of the cell cycle.STUB1TPR repeat; Zn-finger, modified RING8.00.06TRIP4Transcription coactivator of nuclear7.60.02receptors; plays a pivotal role in thetransactivation of NF-kappa-B, SRF, AP1.WNT5BLigand for members of the frizzled family7.50.04of seven transmembrane receptors.Probable developmental protein.TXNLThioredoxin-like protein 1; 32 kDa7.50.03thioredoxin-related proteinARRB1Beta-arrestin: regulates beta-adrenergic7.30.06receptor function.HNRPH2Component of the heterogenous nuclear7.30.04ribonucleoprotein (hnRNP) complexGSTZ1Bifunctional enzyme with minimal7.20.05glutathione-conjugating activity and lowglutathione peroxidase activityPKP4May play a role in junctional plaques7.10.05CD84leukocyte antigen CD84 (leukocyte7.00.05antigen)TUBGCP6Gamma-tubulin complex is necessary for6.90.02microtublule nucleation at the centrosomeQ9NUU6transmembrane6.90.05VAMP5May participate in trafficking events that6.80.05are associated with myogenesisRNF41Zn-finger, RING6.70.06ENSG00000178042ATP/GTP-binding site motif A (P-loop);6.70.04Zn-finger, C2H2 subtypeTSPAN2May play a role in signaling inoligodendrocytes in the early stages of their6.60.04terminal differentiationQ96E44Beta and gamma crystallin; Nuclear protein6.50.03SETQ8NHE2SF21 protein6.40.05PIP5K1BPhosphatidylinositol-4-phosphate 5-kinase6.40.03U2AF1L2U2 small nuclear ribonucleoprotein6.30.06auxiliary factor 35 kDa subunit related-protein 1EIF2C4Plays an important role in the eukaryotic6.30.03peptide chain initiation processDACH2Bipartite nuclear localization signal;6.20.02Transforming protein SkiQ7Z620C2 domain6.20.06Q96IZ9PTD016 protein.6.00.04IFNA2Interferon-alpha; produced by6.00.04macrophages, with antiviral activities.CCSDelivers copper to copper zinc superoxide6.00.04dismutase (SOD1)Q8TDS9putative G-protein coupled receptor6.00.03GPCR42.SPG6WW/Rsp5/WWP domain5.80.05TIRAPAdapter involved in the TLR4 signaling5.70.04pathway in the innate immune response.Acts via IRAK2 and TRAF-6, leading tothe activation of NF-kappa-B, MAPK1,MAPK3 and INK, resulting in cytokinesecretion and the inflammatory responseQ96MA7coiled-coil; low complexity5.70.06UBXD2UBX domain-containing protein 25.60.04ANKMY1Ankyrin repeat and MYND domain protein5.60.011; Testis-specific ankyrin-like protein 1;PCCBPropionyl-CoA carboxylase beta chain,5.60.05mitochondrial precursor;TNFSF5IP1Hepatocellular carcinoma susceptibility5.60.05protein; x 003 protein; TNF superfamily,member 5-induced protein 1PAK2The activated kinase phosphorylates5.50.05ribosomal protein S6, histone H4 andmyelin basic proteinWdr68WD-repeat protein 68; WD-repeat protein5.40.03An11 homologSMYD3Histone methyltransferase. Important role5.30.04in transcriptional activationCENTB2GTPase-activating protein for the ADP5.30.02ribosylation factor familyTIMM9Likely involved in the import and insertion5.20.05of hydrophobic membrane proteins into themitochondrial inner membraneQ9P1G1signal peptide5.20.05C21orf108Nucleolar preribosomal-assoc. protein 15.10.04FBXW5Cyclin-like F-box; G-protein beta WD-405.00.02repeatARHGAP20RA domain; RhoGAP domain4.90.04SIRT1NAD-dependent deacetylase, which4.90.05regulates processes such as apoptosis andmuscle differentiationC13orf1SPla/RYanodine receptor SPRY4.90.03REPS1May coordinate the cellular actions of4.90.02activated EGF receptors and Ral-GTPasesBACH1Transcriptional regulator that acts as4.80.02repressor or activator. Binds, to NF-E2binding sites. Coordinates transcriptionactivation/repression by MAFKPPP1R13BRegulator that plays a central role in4.80.03regulation of apoptosis via its interactionwith p53/TP53.PLA1AEsterase/lipase/thioesterase, active site;4.80.03ARF1GTP-binding protein; allosteric activator4.70.04involved in protein trafficking amongdifferent compartments.CNTN6ABC transporter; Fibronectin, type III;4.60.04Immunoglobulin-likeDHRS2May inhibit cell replication4.60.05SYT1May have regulatory role in the membrane4.50.02interactions during traffickingQ8NG51Zn-finger, Ran-binding4.40.03EBF3Transcriptional activator which recognizes4.40.05variations of the palindromic sequence“ATTCCCNNGGGAATT”ITPKBInositol-trisphosphate 3-kinase B4.30.03TCN1Vitamin B12-binding protein. Transports4.30.02cobalamin into cellsSULT1C1Catalyzes the sulfate conjugation of many4.30.05drugs, xenobiotic compounds, hormones,and neurotransmitters.LRRN1Leucine-rich repeat protein with cysteine-4.20.05rich flanking C-terminal region;Fibronectin, type III; Immunoglobulin-like;MINK1; MAP4K6Serine/threonine kinase that may play a4.10.04role in the response to environmentalstress. Acts upstream of JUN.DBIFunctions as an intracellular carrier of acyl-4.10.04CoA esters. May modulate the action of theGABA receptorQ14159ATP/GTP-binding site motif A (P-loop)4.10.02ZHX2Homeobox; Zn-finger, C2H2 type4.10.05COLEC12Protein C2orf4; C21orf19-like protein4.00.03SDCCAG33Zn-finger, C2H2 type4.00.02MPHOSPH6M-phase phosphoprotein 64.00.01Q86TW0Bipartite nuclear localization signal; Zn-4.00.05finger, C-x8-C-x5-C-x3-H typeMFAP1Component of the elastin-associated4.00.03microfibrilsKLRB1C-type lectin4.00.03APOL5May affect the movement of lipids in cyto-4.00.05plasm or allow lipid:organelles bindingGPR30Orphan receptor; possibly for a chemokine3.90.02HYAL4Epidermal growth factor-like domain;3.90.01Glycoside hydrolase, family 56;CCR7Receptor for the MIP3-β chemokine.3.90.02FOXQ1Forkhead box protein Q1; Hepatocyte3.90.04nuclear factor 3 forkhead homolog 1;VBP1Binds specifically to cytosolic chaperonin3.80.01(c-CPN) and transfers target proteins to it.PSMA1Proteasome subunit; a multicatalytic3.80.04proteinase complex that cleaves specificpeptidesKLHDC2Kelch domain containing protein 2;3.80.04Hepatocellular carcinoma-associatedantigen 33; Host cell factor homolog LCPMRPL37Ribosomal protein, mitochondrial, L2.3.80.06mitochondrial ribosomal protein L37ARID3ABinds a VH promoter proximal site3.70.06necessary for induced mu-heavy-chaintranscription.Q8N867unknown3.70.05ASPNAsporin precursor; Periodontal ligament-3.60.06associated protein 1MAP2K4Dual specificity kinase that activates the3.60.05JUN kinases MAPK8 (JNK1) and MAPK9(JNK2) as well as MAPK14 (p38) but notMAPK1 (ERK2) or MAPK3 (ERK1)ELF1Transcription factor that transcriptionally3.60.00activates the LYN and mouse BLKpromotersHIPK2Protein kinase acting as a corepressor of3.50.04several transcription factors, includingSMAD1 and POU4F1/Brn3a and probablyNK homeodomain transcription factors.Inhibits cell growth and promotesapoptosis. Wnt/beta-catenin pathway.RBM3Putative RNA-binding protein 3;3.50.02TMSB10Plays an important role in the organization3.40.05of the cytoskeleton. Binds to and sequestersactin monomers (G actin) and thereforeinhibits actin polymerizationBMPR2Activin type II receptor; Protein kinase;3.40.06TGF-beta receptor/activin receptor, typeI/IIYWHAHAdapter protein implicated in the3.40.02regulation of a large spectrum of bothgeneral and specialized signaling pathways.PPP1CBProtein phosphatase (PP1) essential for cell3.40.01division, glycogen metabolism, musclecontractility and protein synthesis.COX7BNuclear-coded polypeptide chain of3.40.01cytochrome c oxidaseC13orf11coiled-coil; low complexity; signal peptide;3.30.03transmembraneQ9H5A960S Acidic ribosomal protein3.30.02EIF3S12Binds to the 40S ribosome and promotes3.30.01the binding of met-tRNAi and mRNAKPNB1Functions in nuclear protein import, either3.30.06in association with an adapter protein, or asan autonomous nuclear transport receptor.COL5A1Collagen triple helix repeat;3.20.05GSTK1Specific glutathione conjugating activity3.20.05with 1-chloro-2,4-dinitrobenzene (CDNB)ZNF576Zn-finger, C2H2 type3.20.06Q96T82signal peptide; transmembrane3.20.00C14orf132transmembrane3.10.01BAZ2AMay serve a specific role with ISWI in3.10.05maintaining or altering the chromatinstructure of the rDNA locusQ8N2S5Pistil-specific extensin-like protein;3.10.04Proline-rich extensin; Proline-rich regionCBFA2T3Proline-rich region; Zn-finger, MYND type3.10.05MEP1AMeprin A alpha-subunit precursor;3.10.02Endopeptidase-2;MARK3Involved in the specific phosphorylation of3.10.05microtubule-associated proteinsQ8WUC7Bipartite nuclear localization signal3.10.02Q96BW9unknown3.00.04Q9P1E7unknown2.90.05Q8TF23BED finger; Cytochrome c heme-binding2.90.01site; KRAB box; Zn-finger, C2H2 subtypeETNK2Ethanolamine kinase-like protein EKI22.90.04NCK1Adapter protein which associates with2.90.03tyrosine-phosphorylated growth factorreceptors or their cellular substratesHPSEGlycoside hydrolase, family 79, N-terminal2.90.04RCN1May regulate calcium-dependent activities2.90.06in the endoplasmic reticulum lumen orpost-ER compartmentQ92519Protein kinase2.90.02PANK3Plays a role in the physiological regulation2.80.02of the intracellular CoA concentrationBDNFPromotes the survival of neuronal2.80.00populationsQ96EC8Protein of unknown function DUF6492.80.06LY6G6EPutative Ly-6 superfamily member;2.80.02lymphocyte antigen 6 complex, locus G6EMTRF1Mitochondrial peptide chain release factor2.80.04that directs the termination of translationQ9NWD5Bipartite nuclear localization signal2.70.05Q8NBE8BTB/POZ domain; Kelch repeat2.70.02CPEB4RNA-binding region RNP-1 (RNA2.70.04recognition motif)Q86X05Hly-III related proteins2.60.05MLCBImportant role in regulation of both smooth2.60.05muscle & other cell contractile activitySMURF2E3 ubiquitin-protein ligase; interacts with2.60.05SMAD1, SMAD2 and SMAD7 leading toubiquitination and degradation.Q8WVI0low complexity2.60.02ECGF1May have a role in maintaining the2.60.01integrity of the blood vessels. Has growthpromoting activity on endothelial cells,angiogenic activity in vivo and chemotacticactivity on endothelial cells in vitroC20orf107Similar to neuronal thread protein.2.60.04GSK3BParticipates in the Wnt signaling pathway.2.60.04Phosphorylates JUN, thereby reducing itsaffinity for DNAFIGNL1AAA ATPase, central region; ATP/GTP-2.60.00binding site motif A (P-loop)Q9BRJ9Basic helix-loop-helix dimerization domain2.60.04bHLHDACH2Bipartite nuclear localization signal;2.50.02Transforming protein SkiQ9HCE6DH domain2.50.00ATP2B4Mg2+-dependent enzyme catalyzes the2.50.03hydrolysis of ATP coupled Ca2+ effluxQ9H631Mak10 subunit, NatC N(alpha)-terminal2.50.01acetyltransferaseQ9UF01FGF receptor activating protein 1.2.50.05PTSInvolved in the biosynthesis of2.50.03tetrahydrobiopterin, an essential cofactor ofaromatic amino acid hydroxylases.HBG1The epsilon chain is a beta-type chain of2.50.02early mammalian embryonic hemoglobinZNF208KRAB box; Neutral zinc2.40.05metallopeptidases, Zn-finger, C2H2 typeMLL2May be involved in transcriptional2.40.04regulationNPAS2Neuronal PAS domain protein 2; Neuronal2.40.04PAS2; Member of PAS protein 4; MOP4ZNF80Maybe involved in transcript regulation2.40.02Q9GZT3RNA-binding region RNP-1 (RNA2.40.06recognition motif)Q8N1Q6coiled-coil; low complexity2.40.04INSIG1May play a role in growth & differentiation2.40.03of tissues involved in metabolic control.DDX39ATP/GTP-binding site motif A (P-loop);2.30.05DEAD/DEAH box helicase;Q8NG48WINS1 protein isoform 1.2.30.03SERPINB8Serpin B8; Cytoplasmic antiproteinase 2;2.30.00CAP2; CAP-2; Protease inhibitor 8CDC2L1Appears to play multiple roles in cell cycle2.30.01progression, cytokinesis and apoptosis.NCAM2May play important roles in selective2.30.02fasciculationEMDEmerin2.30.01PET112LFormation of correctly charged Gln-2.30.02tRNA(Gln) through transamidation.MBNL1Binds to CUG triplet repeat expansion2.30.05dsRNAHEY2Antifreeze protein, type I; Basic helix-loop-2.30.02helix dimerization domain bHLHADRM1Promotes cell adhesion2.20.02MCL1Involved in programing of differentiation2.20.02and concomitant maintenance of viabilitybut not of proliferation. Isoform 1 inhibitsapoptosis while isoform 2 promotes itNSMAFCouples the p55 TNF-receptor (TNFR1) to2.20.03neutral sphingomyelinase.NDUFB4Transfer of electrons from NADH to the2.20.02respiratory chain.VPREB1Associates with the Ig-mu chain to form a2.20.05molecular complex that is expressed on thesurface of pre-B-cells. Regulates ig generearrangements in the early steps of B-celldifferentiationEIF3S7Binds to the 40S ribosome and promotes2.20.05the binding of methionyl-tRNAi andmRNA. Associates with the subunit p170of eIF-3KIAA1404Protein KIAA14042.20.04HILS1Implicated in chromatin remodeling and/or2.20.04transcriptional regulation duringspermiogenesis, the process of spermatidmaturation into spermatozoaHSD3B1Crucial role in the biosynthesis of all2.20.02classes of hormonal steroids; 3beta-HSD isa bifunctional enzyme, that catalyzes theoxidative conversion of some steroids andthe oxidative conversion of ketosteroids.Q8IVU3Regulator of chromosome condensation,2.20.03HECT (Ubiquitin-protein ligase)domainQ8WV60PPR repeat2.20.01DAB1Adapter molecule functioning in neural2.20.02development. May regulate SIAH1 activityICAM3ICAM proteins are ligands for the2.20.05leukocyte adhesion LFA-1 protein (integrinalpha-L/beta-2). ICAM3 is also a ligand forintegrin alpha-D/beta-2WWP2E3 ubiquitin-protein ligase which directly2.20.04transfers the ubiquitin to targeted substratesO14950Calcium-binding EF-hand2.20.04OSGEPL1Glycoprotease (M22) metalloprotease2.20.01RASA2Inhibitory regulator of the Ras-cyclic AMP2.20.02pathway. Binds inositol tetrakisphosphateMAFFInteracts with the upstream promoter2.10.04region of the oxytocin receptor gene. Maybe a transcriptional enhancer involved inthe cellular stress responseSNAPAPMay have a role in the mechanisms of2.10.05SNARE-mediated membrane fusion innon-neuronal cellsGNA15Guanine nucleotide-binding protein (G2.10.05proteins) possibly involved as modulator ortransducers in transmembrane signalingCASRSenses changes in the extracellular2.10.02concentration of calcium ions. The activityof this receptor is mediated by a G-proteinthat activates a phosphatidylinositol-calcium second messenger systemPOLR2DDNA-dependent RNA polymerase2.10.05Associates with POLR2GQ9UPS8Ankyrin; RepA/Rep+ protein KID2.10.01FER1L3May play a role in membrane regeneration2.10.02and repairCCNB1IP1E3 ubiquitin ligase. Modulates cyclin B2.10.03levels. Overexpression causes delayedentry into mitosisGCN5L1Biogenesis of lysosome-related organelles2.10.04complex-1, subunit 1;BTBD2BTB/POZ domain containing protein 22.10.04EHHADHeIF-2 functions in the early steps of protein2.10.00synthesis by forming a ternary complexwith GTP and initiator tRNA.Q96MX1down-regulated by Ctnnb1, a.2.10.01NDUFV1Transfer of electrons from NADH to the2.10.01respiratory chain.Q9NZY8unknown2.10.05CD81May play an important role in the2.10.06regulation of lymphoma cell growth.Possibly involved in signal transduction.May acts as the viral receptor for HCVPAIP2PABP-interacting protein 2; polyA-binding2.10.06protein-interacting protein 2.ATP6V1G2Catalytic subunit of the peripheral V12.10.04complex of vacuolar ATPase. Responsiblefor acidifying a variety of intracellularcompartments in eukaryotic cellsCOL9A2Structural component of hyaline cartilage2.00.02and vitreous of the eyeFBXW2Substrate-recognition component of the2.00.02SCF (SKP1-CUL1-F-box protein)-type E3ubiquitin ligase complexGALK1Major enzyme for galactose metabolism2.00.06O15069Nascent polypeptide-associated complex2.00.05NACZNF140ATP/GTP-binding site motif A (P-loop);2.00.04KRAB box; Zn-finger, C2H2 typeWFS1Wolframin2.00.03ENSG00000106603signal peptide; transmembrane2.00.03LECT2Neutrophil chemotactic activity. Positive2.00.01regulator of chondrocyte proliferationQ8IYT6Bipartite nuclear localization signal2.00.03RBP5Intracellular transport of retinol2.00.04Q8NAC1Protein of unknown function DUF609;2.00.04RNA-binding region RNP-1PCLOMay act as a scaffolding protein2.00.06DBC1Deleted in bladder cancer chromosome2.00.01region candidate 1.STMN2May play a role in neuronal differentiation,2.00.02and in modulating membrane interactionwith the cytoskeletonPANK2Maybe the master regulator of the CoA2.00.02biosynthesisQ9NVS3IQ calmodulin-binding region2.00.02ARHGDIARegulates the GDP/GTP exchange reaction2.00.03of Rho proteinsQ9H607Bipartite nuclear localization signal1.90.03Q9P233Calponin-like actin-binding; Eggshell1.90.04protein; Leucine-rich repeatDPYSDihydropyrimidinase; Hydantoinase; DHP1.90.05BTBD12BTB/POZ domain1.90.03PELI1Scaffold protein involved in the IL-11.90.00signaling pathway via its interaction withthe complex containing IRAK kinases andTRAF6. Required for NFκ-B activation &IL-8 gene expression in response to IL-1TTLL3Tubulin tyrosine ligase-like protein 3;1.90.01HOTTLHDCHistidine decarboxylase; HDC1.90.04NOTCH4Class II histocompatibility antigen, beta1.90.00chain, beta-1 domain;TSPAN9Tetraspanin-9; Tspan-9; Tetraspan NET-51.90.01Q8TBL3NULL1.90.04Q96NJ4signal peptide1.90.05CD151Essential for proper assembly of the1.90.01glomerular & tubular basement membranesin kidneyUSP45Ubiquitin thiolesterase, family 2; Zn-finger1.90.04in ubiquitin thiolesteraseSLC30A5Cation efflux protein1.90.05SIAT4CMay catalyze the formation of the sugar1.90.00sequences found in terminal carbohydrategroups of glycoproteins and glycolipids.GBAGlucosylceramidase precursor; Beta-1.90.03glucocerebrosidase; Acid beta-glucosidase;AKAP6Binds to type II regulatory subunits of1.90.06protein kinase A and anchors/targets themto the nuclear membrane or sarcoplasmicreticulum. May act as an adapter forassembling multiprotein complexesQ96LI1coiled-coil; low complexity1.90.02Q9Y4C1Transcription factor jumonji, jmjC1.90.06FKBP11PPIases accelerate the folding of proteins1.80.00during protein synthesisC12orf4low complexity1.80.01Q7Z5J8Ankyrin; Armadillo repeat; RNA-binding1.80.03region RNP-1 (RNA recognition motif)Q8NEQ3unknown1.80.02MAP2K6Catalyzes the concomitant phosphorylation1.80.01of a threonine and a tyrosine residue inMAP kinase p38 exclusivelyCRIM1Antistasin; Eukaryotic thiol (cysteine)1.80.04protease;MUC1Mucin, May play a role in adhesive1.80.01functions and in cell-cell interactions,metastasis and signaling. Possible protectivelayer on epithelial surfaces. Direct orindirect interaction with actin cytoskeletonF13A1Factor XIII stabilizes fibrin clots. Also1.80.02cross-link alpha-2-plasmin inhibitor, orfibronectin, to the alpha chains of fibrinMRPS28Mitochondrial 28S ribosomal protein S28;1.80.05S28mt; MRP-S28; MRP-S35FYBActs as an adapter protein of the FYN and1.80.03SH2-domain-containing leukocyte protein-76 (SLP76) signaling cascades in T cells.Modulates the expression of interleukin-2TFR2Mediates cellular uptake of transferrin-1.80.01bound iron in a non-iron dependentmanner.Q96PY3Leucine-rich repeat1.80.03TRAPPC3May play a role in vesicular transport from1.80.04endoplasmic reticulum to GolgiQ9NUQ9NULL1.80.04O60844Jacalin-related lectin1.80.02PTPRFPossible cell adhesion receptor; has intrinsic1.80.00protein tyrosine phosphatase activitySOX6Binds specifically to the DNA sequence1.80.02“AACAAT-3”RAGEAble to phosphorylate several exogenous1.80.03substrates & undergo autophosphorylationPLAURReceptor for urokinase plasminogen1.80.01activator. Plays a role in localizing andpromoting plasmin formation.ENTPD5Likely to promote reglycosylation reactions1.80.02involved in glycoprotein folding & qualitycontrol in the endoplasmic reticulum.UNC5BATP/GTP-binding site motif A (P-loop);1.80.04Death domain; Immunoglobulin-likeCHCHD3Protein of unknown function DUF7371.80.03LMNALamins are components of the nuclear1.80.05lamina, a fibrous layer on thenucleoplasmic side of the inner nuclearmembrane that is thought to provide aframework for the nuclear envelopeZNF254May function as a transcription factor1.80.03POLR1BRNA polymerase beta subunit1.80.02RUFY2Cytochrome c heme-binding site;1.80.06CDC34Catalyzes the covalent attachment of1.80.04ubiquitin to other proteinsDEF6Calcium-binding EF-hand; Pleckstrin-like;1.80.00TropomyosinPKHD1L1ATP/GTP-binding site motif A (P-loop);1.80.05Cell surface receptor IPT/TIGFGL1Has hepatocyte mitogenic activity1.80.01TRPC7Thought to form a receptor-activated non-1.80.05selective calcium permeant cation channel.GLULGlutamine synthetase;1.80.00IGHG3Ig alpha is the major immunoglobulin class1.80.01in body secretions. It may serve both todefend against local infection and toprevent access of foreign antigens to thegeneral immunologic systemDSPG3May have a role in bone formation and also1.80.03in establishing the ordered structure ofcartilage through matrix organizationC10orf82unknown1.80.05ENSG00000144872Ribosomal protein L39e1.70.01FRMD1Band 4.1 domain1.70.01Q96J64Somatomedin B; Thrombospondin, type I1.70.05PCBDInvolved in tetrahydrobiopterin1.70.01biosynthesis.INPP4AInositol polyphosphate-4-phosphatase I1.70.04TRIM39Tripartite motif protein 39; RING finger1.70.04protein 23; Testis-abundant finger proteinAKAP11Binds to type II regulatory subunits of1.70.03protein kinase A and anchors/targets themZNF292May function as a transcription factor1.70.04APG10LAutophagocytosis associated protein, C-1.70.00terminalZNF177May be involved in transcriptional1.70.05regulationTSHRReceptor for thyrothropin. Plays a central1.70.01role in controlling thyroid cell metabolism.EAF1Proline-rich extensin; Proline-rich region1.70.02Q8N2H5Adrenodoxin reductase; Regulator of1.70.02chromosome condensationETV4Ets-domain; PEA3-type ETS-domain1.70.01transcription factor, N-terminalACE2Neutral zinc metallopeptidase1.70.05NTRK3Tyrosine-protein kinase receptor for1.70.00neurotrophin-3 (NT-3). Known substratesfor trk receptors are SHC1, PI-3 kinase,and PLCG1.FADS1Cytochrome b5; Fatty acid desaturase1.70.01familyQ9H679low complexity; signal peptide;1.70.01transmembraneRBMS3Paraneoplastic encephalomyelitis antigen;1.70.01RNA-binding region RNP-1C20orf173Hypothetical protein C20orf1731.70.06Q9BY88Calcium-binding EF-hand1.70.01USP6NLRabGAP/TBC domain1.70.04NDUFB7Transfer of electrons from NADH to the1.70.03respiratory chain.FRZBSoluble frizzled-related proteins (sFRPS)1.70.01function as modulators of Wnt signaling.They have a role in regulating cell growthand differentiation in specific cell types.Q9NTD9unknown1.70.04ZFP36Probable regulatory protein with a novel1.70.05zinc finger structure involved in regulatingthe response to growth factors.PPM1DRequired for the relief of p53-dependent1.70.03checkpoint mediated cell cycle arrest.Q8NBM8NULL1.70.01STAT1Signal transducer and activator of1.70.04transcription that mediates signaling byinterferons (IFNs).DSC1Component of intercellular desmosome1.70.01junctions. Involved in the interaction ofplaque proteins and intermediate filamentsmediating cell-cell adhesion.GMEB2Trans-acting factor that binds to1.70.05glucocorticoid modulatory elements. Bindsalso to the transferrin receptor promoter.PPICCatalyzes the cis-trans isomerization of1.70.03proline imidic peptide bonds in oligo-peptides to accelerate protein foldingARRDC2Arrestin1.70.01ADPRHL1ADP-ribosylglycohydrolase1.70.01HES7Basic helix-loop-helix dimerization domain1.70.00bHLH; Proline-rich extensin;Wdr68; Han11WD-repeat protein 68; WD-repeat protein1.70.02An11 homologPSMB7Proteasome subunit1.70.01SLC25A20Transport of acylcarnitines of different1.60.05length across the mitochondrial innermembrane for their oxidationCACNA2D2Cache domain; von Willebrand factor, type A1.60.05EDG2Receptor for lysophosphatidic acid (LPA),1.60.00a mediator of diverse cellular activities.Coupled to heteromeric G proteinsTFECBasic helix-loop-helix dimerization domain1.60.04bHLHETF1Directs the termination of nascent peptide1.60.01synthesis (translation)NPPurine nucleoside phosphorylase; Inosine1.60.00phosphorylase; PNPSLC11A1Divalent transition metal (iron and1.60.04manganese) transporter involved in ironmetabolism and host resistance to certainpathogens. Controls natural resistance toinfection with intracellular parasites.Q8NDJ4TBP-interacting 1201.60.02TRPV1Ankyrin; Ion transport protein1.60.05CORO1AMay be a crucial component of the1.60.03cytoskeleton of highly motile cellsCRHBPBinds CRF and inactivates it. May prevent1.60.04inappropriate pituitary-adrenal stimulationin pregnancyRPO2TC1; PC4General coactivator that functions1.60.00cooperatively with TAFs and mediatesfunctional interactions between upstreamactivators and the general transcriptionalmachinery. Binds single-stranded DNAFMNL2Actin-binding FH2; Proline-rich extensin;1.60.05Wilm's tumour proteinFZD10Receptor for Wnt proteins. May be1.60.05involved in transduction and intercellulartransmission of polarity information duringtissue morphogenesis and/or indifferentiated tissuesMAN1C1Involved in the maturation of Asn-linked1.60.02oligosaccharides.PLAC8Placenta-specific gene 8 protein; C151.60.04proteinC6orf211Protein of unknown function DUF891.60.01PTPN18Differentially dephosphorylate auto-1.60.00phosphorylated tyrosine kinases which areknown to be overexpressed in tumor tissuesLRP11Low density lipoprotein-receptor, class A1.60.05ITPAHydrolyzes ITP and dITP to their1.60.06respective monophosphate derivatives.May be the major enzyme responsible forregulating ITP concentration in cellsCOG3Involved in ER-Golgi transport1.60.00CFL2Controls reversibly actin polymerization1.60.02and depolymerization in a pH-sensitivemanner. Major component of intranuclearand cytoplasmic actin rodsQ8N5X0low complexity; signal peptide;1.60.01transmembraneSIAT8BMay transfer sialic acid to N-linked1.60.03oligosaccharides of glycoproteinsVARS2Valyl-tRNA synthetase1.60.01RAB13Could participate in polarized transport, in1.60.05assembly and/or activity of tight junctionsTGIFActive transcriptional corepressor of1.60.02SMAD2. May participate in thetransmission of nuclear signals duringdevelopment and in the adultKIF13APlus end-directed microtubule-dependent1.60.04motor protein involved in mannnose-6-phosphate receptor transport to the plasmamembraneRGS17Inhibits signal transduction by increasing1.60.03the GTPase activity of G protein alphasubunits thereby driving them into theirinactive GDP-bound formENSG00000180649RNA-binding region RNP-1 (RNA1.60.05recognition motif)Q9UFK2low complexity; signal peptide;1.60.05transmembraneQ9Y6U7Proline-rich region; Zn-finger, RING1.60.05NOL4Nucleolar protein 41.60.05PTBP1Plays a role in pre-mRNA splicing. Binds1.60.01to the polypyrimidine tract of introns.Q9NW32low complexity1.60.01RTTNRotatin.1.60.06CDCA4May be involved in molecular regulation of1.60.01hematopoietic stem cells and progenitorcell lineage commitment and differentiationTBC1D14RabGAP/TBC domain1.60.01Q9H693low complexity1.60.00RGS11Inhibits signal transduction by increasing1.60.05the GTPase activity of G protein alphasubunits thereby driving them into theirinactive GDP-bound formGIMAP2;GTPase, IMAP family member 2;1.60.01IMAP2Immunity-associated protein 2; hIMAP2SSBP3May be involved in transcription regulation1.60.06of the alpha 2(I) collagen gene where itbinds to the single-stranded polypyrimidinesequences in the promoter regionEIF4EL3Recognizes and binds the 7-1.60.01methylguanosine-containing mRNA capduring an early step in the initiation ofprotein synthesis and facilitates ribosomebinding by inducing the unwinding of themRNAs secondary structuresMNDAMay act as a transcriptional1.60.04activator/repressor in the myeloid lineage.Plays a role in the granulocyte/monocytecell-specific response to interferon.Stimulates the DNA binding of thetranscriptional repressor protein YY1O95053low complexity1.60.01GNA13Guanine nucleotide-binding proteins (G1.60.02proteins) are involved as modulators ortransducers in various transmembranesignaling systemsC13orf17Protein of unknown function DUF2981.60.02EXOSC4Component of exosome:exoribonuclease1.60.01complex. Required for the processing ofthe 7S pre-RNA to the mature 5.8S rRNA.Has an exonuclease activityO76052low complexity1.60.03F11RSeems to plays a role in epithelial tight1.50.05junction formation. Plays a role inregulating monocyte transmigrationinvolved in integrity of epithelial barrier.Involved in platelet activationSLC35B3CGI-19 protein; solute carrier family 35,1.50.04member B3Q8WVP6low complexity1.50.05Q9NYY8NULL1.50.01SLC9A7Na+/H+ exchanger, isoform 6 (NHE6);1.50.06Sodium/hydrogen exchanger;FUT1Creates a soluble precursor oligosaccharide1.50.02FuC-α(1,2)Galβ-called the H antigenDOT1LHistone methyltransferase. Methylates Lys-1.50.0479 of histone H3. Nucleosomes arepreferred as substrates, cf. free histonesPPP2R2CThe B regulatory subunit might modulate1.50.02substrate selectivity and catalytic activity,KIAA2010EVH1; Protein of unknown function1.50.05DUF625Q9BU62Similar to DNA segment, Chr 17, human1.50.02D6S51E.PPP2CAPP2A can modulate the activity of1.50.05phosphorylase B kinase casein kinase 2,mitogen-stimulated S6 kinase, and MAP-2kinase.SLC22A11General substrate transporter1.50.04MAN2B1Necessary for the catabolism of N-linked1.50.02carbohydrates released during glycoproteinturnover. Cleaves all known types of alpha-mannosidic linkagesO94940SAM (and some other nucleotide) binding1.50.04motifAPOBEC3GAPOBEC-like, C-terminal; Cytidine/1.50.03deoxycytidylate deaminase,Q14843Calcium-binding EF-hand1.50.04Q9P1G3Protein of unknown function DUF1851.50.06KCNN3Voltage-independent potassium channel1.50.01activated by intracellular calcium.Activation is followed by membranehyperpolarization.HSD17B12Glucose/ribitol dehydrogenase; Short-chain1.50.02dehydrogenase/reductase SDRZNF83May be involved in transcriptional1.50.02regulationSIAT8EMay be involved in the synthesis of1.50.02gangliosides GD1c, GT1a, GQ1b and GT3PHLDB2Pleckstrin-like1.50.01Q9H2V5Bipartite nuclear localization signal1.50.04AKAP13Stimulates exchange activity on Rho1.50.01proteins in vitroCRHR1Receptor for corticotropin releasing factor.1.50.01Q9BVM2DPCD protein.1.50.01HOXD3Sequence-specific transcription factor, part1.50.00of a developmental regulatory systemHDAC9Responsible for the deacetylation of lysine1.50.01residues on the N-terminal part of the corehistones (H2A, H2B, H3 and H4).HV2G_HUMANIg heavy chain V-II region NEWM1.50.04ZNF514KRAB box; Zn-finger, C2H2 subtype;1.50.00CPS1Involved in the urea cycle in removing1.50.03excess ammonia from the cellTTC12Tetratricopeptide repeat protein 12 TPR1.50.01PLXNA3Putative receptor involved in the1.50.00development of neural & epithelial tissuesCFLARApoptosis regulator protein which may1.50.01function as a crucial link between cellsurvival and cell death pathways inmammalian cells. Inhibitor of TNFRSF6mediated apoptosis.TCBA1T-cell lymphoma breakpoint-associated1.50.01target 1.RCE1Proteolytically removes the C-terminal1.50.00three residues of farnesylated andgeranylated proteins. Processes K-Ras, N-Ras, H-Ras, RAP1B and G-gamma-1PLA2G5PA2 catalyzes the calcium-dependent−1.50.02hydrolysis of the 2-acyl groups in 3-sn-phosphoglycerides. May be involved in theproduction of lung surfactantSTX16SNARE involved in a vesicular transport−1.50.00step within the Golgi stackTGM5Catalyzes the cross-linking of proteins and−1.50.00the conjugation of polyamines to proteins.Q8N7V2low complexity−1.50.00SLC35F3Solute carrier family 35, member F3.−1.50.04Q96LR7unknown−1.50.01HRMT1L3Probably methylates the guanidino−1.50.04nitrogens of Arg residues in some proteinsQ9NUJ7Phosphatidylinositol-specific−1.50.03phospholipase C, X domainSLC1A6Transports L-glutamate and also L- and D-−1.50.05aspartate, in symport with Na+KIAA1024UPF0258 protein KIAA1024−1.50.04MAML3CAG repeat containing (glia-derived nexin−1.50.00I alpha);FMO5In contrast with other forms of FMO it does−1.50.01not seem to be a drug-metabolizing enzymeQ9BTD3hole protein.−1.50.00COL5A2Collagen type II specific for cartilaginous−1.50.02tissues. It is a minor connective tissuecomponent of nearly ubiquitousdistribution.CD48Ligand for CD2. Might facilitate inter-−1.50.03action between activated lymphocytes.Probably involved in regulating T-cellactivationNDUFAF1Chaperone protein involved in assembly of−1.50.05the mitochondrial NADH:ubiquinoneoxidoreductase complexQ9NWV6Protein kinase−1.50.02Q9C0D5ATP/GTP-binding site motif A (P-loop);−1.50.01Ankyrin; TPR repeatTRIM11Tripartite motif protein 17; Testis RING−1.50.05finger protein; RING finger protein 16ENSG00000187700low complexity−1.50.04Q9H7M9Immunoglobulin-like−1.50.04IL12ACytokine that can act as a growth factor for−1.50.03activated T and NK cells, enhance the lyticactivity of NK/lymphokine-activated Killercells, and stimulate the production of IFN-gamma by resting PBMCQ8IZC1Protein of unknown function DUF634−1.60.03GRSF1Binds RNAs containing the 14 base G-rich−1.60.03elementNBR2Protein NBR2; Next to BRCA1 gene 2−1.60.02proteinQ8TF23BED finger; Cytochrome c heme-binding−1.60.01site; KRAB box; Zn-finger, C2H2 subtype;ZNF78L1May function as a transcription factor−1.60.01ENSG00000184319Bipartite nuclear localization signal;−1.60.01Ribosomal L23 protein; Ribosomal proteinL23, N-terminal domainARID1AInvolved in transcriptional activation and−1.60.01repression of select genes by chromatinremodelingPRKWNK4Regulates the activity of the thiazide-−1.60.02sensitive Na—Cl cotransporter, SLC12A3,by phosphorylationEXTL2Glycosyltransferase required for the−1.60.06biosynthesis of heparan-sulfateASPHAspartyl/Asparaginyl beta-hydroxylase−1.60.02Q9NV64low complexity; transmembrane−1.60.01Q96MY4low complexity−1.60.02ADAM30May be involved in spermatogenesis and−1.60.03fertilizationEFHD1EF-hand domain-containing protein 1;−1.60.02Swiprosin-2SGTBSmall glutamine-rich tetratricopeptide−1.60.03repeat-containing protein B;Q9H0J1low complexity; transmembrane−1.60.03ENSG00000185305ATP/GTP-binding site motif A (P-loop)−1.60.03A2BP1Ataxin-2-binding protein 1−1.60.06Q86XE5Dihydrodipicolinate synthetase−1.60.03FOXK2May be involved in both positive and−1.60.03negative regulation of important viral andcellular promoter elementsNFIARecognizes and binds the palindromic−1.60.04sequence “TTGGCNNNNNGCCAA”;present in viral and cellular promoters.ITGA9Integrin alpha-9/beta-1 is a receptor for−1.60.03VCAM1, cytotactin and osteopontin.ZCCHC7Zn-finger, CCHC type−1.60.00DOCK1Involved in cytoskeletal rearrangements−1.60.00required for phagocytosis of apoptotic cellsand cell motility. Functions as a guaninenucleotide exchange factor (GEF), whichactivates Rac Rho small GTPases.ENSG00000186409coiled-coil; low complexity−1.60.01Q8IXL9IQ calmodulin-binding region−1.60.03EIF2AK3Phosphorylates the alpha subunit of−1.60.03eukaryotic translation-initiation factor 2(EIF2)Q8NDC9Initiation factor 2B−1.60.04SH3BGRSH3 domain-binding glutamic acid-rich−1.60.03protein;CDY1Testis-specific chromodomain protein Y 1−1.60.03Q8TAZ0Ubiquitin system component Cue−1.60.01REV1LUMUC-like DNA-repair protein−1.70.02Q96IR7unknown−1.70.06SMCR8Smith-Magenis syndrome chromosome−1.70.02region, candidate 8.ABCA6ABC transporter; ATP/GTP-binding site−1.70.02motif A (P-loop)NRIP1Modulates transcriptional activation by−1.70.01steroid receptors such as NR3C1, NR3C2and ESR1. Also modulates transcriptionalrepression by nuclear hormone receptorsO95792Zn-finger, A20-like; Zn-finger, AN1-like−1.70.03ITGA5Integrin alpha-5/beta-1 is a receptor for−1.70.05fibronectin & fibrinogen; recognizes RGDRPL10A60S ribosomal protein L10a−1.70.05PTPRBReceptor-type tyrosine-protein phosphatase−1.70.02beta precursor; Protein-tyrosinephosphatase beta; R-PTP-betaADCY9May play a fundamental role in situations−1.70.02where interplay between intracellular Ca2+and cAMP determines the cellular function.AKAP13Stimulates exchange activity on Rho−1.70.05proteins in vitro, but not on CDC42, Ras orRac. May bind calcium ionsO94948RUN domain−1.70.05STK39May act as a mediator of stress-activated−1.70.01signalsTFB1MImmunoglobulin/major histocompatibility−1.70.05complex; Ribosomal RNA adeninedimethylaseNJMU_HUMANMay have a role in spermatogenesis−1.70.05Q8N4S7Hly-III related proteins−1.70.03ENSG00000166737low complexity; transmembrane−1.70.01Q8WWN8Human Rev interacting-like protein (hRIP);−1.70.02Pleckstrin-like; RA domain; RhoGAPdomain; Sterile alpha motif SAMQ9Y2M0Bipartite nuclear localization signal−1.70.01Q9BXY2Protein kinase; Tyrosine protein kinase−1.80.03TRPM4Ion transport protein−1.80.04NALP1Able to form cytoplasmic structures termed−1.80.04death effector filaments. Enhances APAF1and cytochrome c-dependent activation ofpro-caspase-9 and consecutive apoptosis.NAALAD2Has N-acetylated-alpha-linked-acidic−1.80.01dipeptidase (NAALADase) activity.GPR55Rhodopsin-like GPCR superfamily−1.80.00DKK1Inhibitor of Wnt signaling pathway−1.80.01GAS1Specific growth arrest protein involved in−1.80.01growth suppression.PCDH9Potential calcium-dependent cell-adhesion−1.90.01proteinAPBA3May modulate processing of the β-amyloid−1.90.05precursor protein (APP) and henceformation of beta-APPWBSCR19Bipartite nuclear localization signal;−1.90.01ConnexinsPPFIA3Sterile alpha motif SAM−1.90.03UPK2Component of the asymmetric unit−1.90.01membrane (AUM); a highly specializedbiomembrane elaborated by terminallydifferentiated urothelial cells.TFCP2L3CP2 transcription factor−1.90.01Q9H8Y5Ankyrin; Prenyl group binding site (CAAX−1.90.03box); Zn-finger, C2H2 typeADHFE1Iron-containing alcohol dehydrogenase−1.90.01O14804Aminotransferase, class-II; Beta-ketoacyl−1.90.02synthase; G-protein coupled receptorsfamily 2 (secretin-like);ZNF208May function as a transcription factor−2.00.00Q9H5D6low complexity−2.00.01TMF1This protein binds the HIV-1 TATA−2.10.01element and inhibits transcriptionalactivation by the TATA-binding proteinSGCGComponent of the sarcoglycan complex,−2.10.01which forms a link between the F-actincytoskeleton and the extracellular matrixKLHL21;Kelch-like protein 21−2.20.06ADRA2AAlpha-2 adrenergic receptors mediate the−2.20.04catecholamine-induced inhibition ofadenylate cyclase through the action of Gproteins.ENSG00000140876NUDIX hydrolase−2.20.06SHANK1Seems to be an adapter protein that may−2.20.04play a role in the structural and functionalorganization of the dendritic spine andsynaptic junctionQ8IY68low complexity−2.30.01NR4A1Orphan nuclear receptor−4.20.05


Bioinformatic analysis of the microarray data by supervised clustering of the differentially expressed genes induced by the peptide, followed by mapping of the responses to signal transduction pathways indicated that the synthetic peptide can potentially induce signaling, and activate JAK (Janus Kinases) and the STAT (signal transducers and activators of transcription) family of transcription factors. It had been previously described that the human host defense peptide LL-37 activates MAP kinases (FIG. 4) but the JAK-STAT pathway had not been implicated. These microarray data showed evidence that SEQ ID NO: 7 has the potential to differentially induce transcriptional responses of the genes upstream and downstream of the JAK and STAT family of transcription factors (FIG. 13), as well as activate PI3-Kinase an important modulator of extracellular signals. The peptide (SEQ ID NO:7) also significantly up-regulated the expression regulators of NFκB pathway e.g. TIRAP and NFκB2. This observation reveals a mechanism for the intervention of host defense peptides in immune responses.


Transcriptional analysis of responses induced by the synthetic peptide using qPCR showed that the peptide was able to induce the expression of chemokines and interleukins such as MCP-3, MCP-1, CXCL-1, and IL-6, IL-19 (IL-10 superfamily) and IL-8, all of which are critical in cell recruitment and movement (FIG. 14). rMCP-1 is known to protect against Salmonella and Pseudomonas infections when given 6 hours prior to the initiation of infection in mice (Infection and Immunity 62:377-383, 1994), and to protect against acute septic peritonitis sue to bowel perforation (Journal of Immunology 163: 6148-54, 1999).


The peptide (SEQ ID NO: 7), as well as LL-37, also induced the expression of IL-19 (FIG. 14) belonging to the IL-1 0 superfamily. It is well known that IL-10 activates the JAK-STAT pathway leading to the up-regulation of the SOCS family members, all of these responses are required to potentially counteract pro-inflammatory responses. Indeed recombinant IL-10 is being considered as a therapeutic for rheumatoid arthritis (Rheum Dis Clin North Am. 24(3):629-39, 1998) and IL-10 protects against endotoxemia (Journal of Experimental Medicine 177:1205-8, 1993) and Pseudomonas-mediated gut-derived sepsis Antimicrobial Agents and Chemotherapy 42:2853-7, 1998).


These genes are thus markers for the biological effects of SEQ ID NO: 7. As JAK-STAT signaling is mediated by members of the IL-10 superfamily, the observation that the synthetic peptide up-regulates the expression of IL-19 further supports the conclusion that one of the signal transduction pathways activated by host defense peptides is the JAK-STAT pathway, resulting in the up-regulation of genes in the SOCS family. The peptide also appears to regulate the NFκB pathway which can in addition lead to the expression of some of the members of the SOCS family. Thus there appears to be subtle changes in the regulation of critical pathways in immune responses (JAK-STAT, NFκB and MAPK) in the presence of the peptide which potentially suggests the mechanism by which the peptide functions in combating pathogenesis. Further analysis of protein production induced by the peptide in human PBMC by ELISA confirmed that SEQ ID NO: 7 is able to induce the secretion of interleukins such as IL-6 and IL-8 in human PBMC within 4 hours of stimulation, but did not induce the expression of the pro-inflammatory cytokine TNFα (FIG. 15). It is thus evident that the peptides induces the expression of critical immunity genes such as chemokines, interleukins, adhesion molecules and transcription factors that are able to modulate specific host defense mechanisms, and thereby prove to be beneficial therapeutics.


In response to bacterial endotoxin (LPS) there were 836 differentially expressed genes in human monocytes (Table 70), and 1012 genes were differentially expressed in response to LPS in the presence of the peptide SEQ ID NO: 7 (Table 71). The peptide appeared to induce the expression of several genes synergistically only in the presence of LPS. These genes are either induced only in the presence of the peptide on LPS stimulation (not with LPS alone) and/or the LPS-induced expression is synergistically enhanced in the presence of the peptide, but not with the peptide alone. These genes included several genes that are critical in immune response in presence of pathogenic challenge such as chemotactic factor CCL20, and CCL23; cytokine receptor EBI3, factors involved in lymphocyte activation such as SLAMF1, CD58, and IL32; regulators of signal transduction such as MAP2K2 (activator of ERK1), DUSP5 (ERK phopsphatase), MAPK8IP3, RIN2 (RAS-effector), RANBP9 (GTP-binding protein that affects Ras-signaling pathway), IP3 3-kinase A,-B3ATF, IRAK3, NM1 (augments cytokine-mediated STAT transcription), SP3, RAP2C, PNRC1, NEK1, CHC1, ZNF219, ZNF593, WIF1, PIM2, CD79A, and LATS2; substrate transporters such as SLC23A3, and SLC17A5; apoptosis regulators such as BOK (Bc1-2 like inhibitor), BIRC3, TNFRSF6, and CASP9; genes associated with plasma membrane such as STIM1, BPAG1, PTPN4, TRIM36, SDK1, and FNDC5 (fibronectin type III); genes involved in selective ion transport and in mediating selective ion-channel such as VGCNL1, TRPC5, CACNA1B, KCNA6, KCNJ2, KCNA10, and AQP9; growth modulating genes and/or those that play a role in wound healing such as FGF10, and AREG; inflammatory mediators such as PTGS2, SOD2, TNFAIP8 and anti-inflammatory gene TNIP3. This indicates that one of the mechanism by which cationic host defense peptides exhibit their protective mechanism is by delicately modulating specific signal pathways only in presence of agonists such as bacterial endotoxin, thus trigger just sufficient amount of inflammation and/or immune responses that is necessary to combat pathogenesis, while also maintaining the anti-inflammatory checks in place in order to prevent excessive inflammation that can lead to sepsis.


Of the 596 genes that were up-regulated by LPS stimulation, 33 genes were suppressed in the presence of the peptide (Table 72), which included the genes for pro-inflammatory TNF-α and NF-κB2 (p52). Interestingly, the peptide alone was able to up-regulate NF-κB2 as analyzed by DNA microarrays, an observation that also has mechanistic implications as individual subunits of NFκB are known to be involved in expression of only a subset of NFκB-regulated genes (the major pair of NFκB subunits p50 and p65 are responsible for much of the pro-inflammatory gene expression). Further transcriptional analysis using qRT-PCR revealed that the peptide exhibited the ability to suppress LPS-induced gene expression of cytokines such as IL-8, IL-6 as well as chemokines such as CXCL-1 within 4 hours of stimulation (FIG. 16). In addition, secretion of LPS-induced pro-inflammatory cytokine TNF-α was suppressed between 30-40% in the presence of peptide SEQ ID NO: 7 in human PBMC (FIG. 17A), as well as in human monocytic cell line THP-1 cells (FIG. 17B). Together, these results suggest that the synthetic peptide not only has the ability to suppress pro-inflammatory responses, e.g. TNF-α induced by LPS, similar to the natural human peptide LL-37 (Mookherjee, et al.), but also may have the potential to activate certain members of the NF-κB transcription factors such as NF-κB2 unlike the natural human peptide

TABLE 70Gene profiling of differentially expressed genes in human monocytes due tobacterial endotoxin (LPS) revealing 836 differentially expressed genesFold changeGene NameGene DescriptionLPSp-studentGPD1Glycerol-3-phosphate dehydrogenase [NAD+],103.40.06cytoplasmic; GPD-C; GPDH-CQ8NI35ATP/GTP-binding site motif A (P-loop);78.40.06PDZ/DHR/GLGF domainQ86Y93ATP/GTP-binding site motif A (P-loop); CSL zinc67.90.05finger; Guanylate-binding protein;KCNK6Exhibits outward rectification in a physiological K(+)61.50.04gradient and mild inward rectification in symmetricalK(+) conditionsRHBDF1Rhomboid-like protein57.30.05FEZ2Involved in axonal outgrowth and fasciculation45.90.05O43348Argininosuccinate synthase43.30.03Q9C098Protein kinase; Serine/Threonine protein kinase43.10.04SMFSMF protein KIAA019441.70.04WBSCR18Williams-Beuren syndrome chromosome region 1836.30.05proteinNRXN1Neuronal cell surface protein that may be involved in cell35.40.05recognition and cell adhesion. May mediate intracellularsignalingDLX5Homeobox protein DLX-534.40.04BNIP1Implicated in the suppression of cell death. Interacts with34.10.04the BCL-2 and adenovirus E1B 19 kDa proteinsQ9P175unknown33.60.03SMURF2E3 ubiquitin-protein ligase which accepts ubiquitin from32.70.05an E2 ubiquitin-conjugating enzyme in the form of athioester and then directly transfers it to targetedsubstrates SMAD1; SMAD2 and SMAD7 to trigger theirubiquitination and degradation.C6orf74Metal-dependent phosphohydrolase, HD region32.60.06PLCG1PLC-gamma is a major substrate for heparin-binding32.60.04growth factor 1 (acidic fibroblast growth factor)-activated tyrosine kinaseLRMPLymphoid-restricted membrane protein.32.00.05CD226Immunoglobulin-like31.10.04Q9Y4T9low complexity30.00.05Q8WUC6Bipartite nuclear localization signal; Class I peptide29.00.05chain release factor domainQ9P278low complexity28.60.03MCART1Mitochondrial carrier triple repeat 127.60.05DKC1Required for ribosome biogenesis and telomere24.20.03maintenance.ENSG00000162701DENN (AEX-3) domain; uDENN domain24.10.05TNCSAM (substrate-adhesion molecule) that appears to23.60.04inhibit cell migration. Ligand for integrins.TRPM8Ion transport protein21.80.05SBNO1Helicase, C-terminal; RNA-binding region RNP-1 (RNA21.50.06recognition motif)Q7RTU0Basic helix-loop-helix dimerization domain bHLH21.40.03CHRNA1After binding acetylcholine, the AChR responds by an21.40.03extensive change in conformation that affects all subunitsand leads to opening of an ion-conducting channel acrossthe plasma membraneKIAA1529coiled-coil; low complexity; transmembrane21.30.06ALDOBFructose-bisphosphate aldolase B; Liver-type aldolase21.00.03Q8NHU6Bipartite nuclear localization signal; Maternal tudor20.90.04proteinQ8N6Q6unknown20.30.01Q9H5P1Zn-finger, C-x8-C-x5-C-x3-H type19.90.06NR2F2Regulation of the apolipoprotein A-I gene transcription.19.00.05Binds to DNA site ASNX13May be involved in several stages of intracellular18.90.06trafficking. Act as a GAP for GalphasTULP2Tubby related protein 2; Tubby-like protein 218.90.05Q96HJ9;unknown18.00.04BLZF1basic leucine zipper nuclear factor 1.17.70.04SLC35A4Nucleotide-sugar transporter17.10.04IDH3GIsocitrate dehydrogenase [NAD] subunit gamma,17.00.05mitochondrial precursor; Isocitric dehydrogenase;NAD(+)-specific ICDHQ9BRK2Protein of unknown function DUF62516.60.03RCL1Plays a role in 40S-ribosomal-subunit biogenesis in the16.20.00early pre-rRNA processing steps at sites A0, A1 and A2that are required for proper maturation of the 18S RNAPMPCBCleaves presequences (transit peptides) from15.60.04mitochondrial protein precursorsO43788;NULL15.20.05GFM2; EFG2;This protein promotes the GTP-dependent translocation15.10.06of the nascent protein chain from the A-site to the P-siteof the ribosomeSYT11May be involved in Ca(2+)-dependent exocytosis of14.80.03secretory vesicles through Ca(2+) and phospholipidbinding to the C2 domain or may serve as Ca(2+) sensorsin the process of vesicular trafficking and exocytosisC10orf11Leucine-rich repeat14.70.05FBN1Fibrillins are structural components of 10-12 nm14.60.05extracellular calcium-binding microfibril. Fibrillin-1-containing microfibrils provide long-term force bearingstructural supportQ9NVK9DH domain; Pleckstrin-like14.50.05MED6Plays a role in transcriptional coactivation14.20.05ENSG00000105849RNA polymerase Rpa43 subunit13.50.04SACM1LSynaptojanin, N-terminal13.50.05SYNE1Involved in the maintenance of nuclear organization and13.40.06structural integrity. Probable anchoring protein whichtethers the nucleus to the cytoskeleton.WNT5BLigand for members of the frizzled family of seven13.20.03transmembrane receptors. Probable developmentalprotein.DHX8Facilitates nuclear export of spliced mRNA by releasing13.00.05the RNA from the spliceosomePRO0461Protein PRO046111.60.03SIRT1NAD-dependent deacetylase, which regulates processes11.50.03such as apoptosis and muscle differentiation bydeacetylating key proteins, including p53, TAF1B, etcNEK2Protein kinase that is involved in mitotic regulation.11.50.04MMP17Endopeptidase that degrades various components of the11.50.06extracellular matrix, such as fibrin. May be involved inthe activation of membrane-bound precursors of growthfactors or inflammatory mediators, such as tumornecrosis factor-alpha.TIMM9Likely to be involved in the import and insertion of11.40.06hydrophobic membrane proteins into the mitochondrialinner membraneARRB1Regulates beta-adrenergic receptor function. Beta-11.10.05arrestins seem to bind phosphorylated beta-adrenergicreceptors, thereby causing a significant impairment oftheir capacity to activate G(S) proteinsO75872rab3 GTPase-activating protein, non-catalytic subunit.11.00.03Q96IZ9PTD016 protein.10.80.03POMT2Transfers mannosyl residues to the hydroxyl group of10.80.05serine or threonine residues.METTL3N6-methyltransferase that methylates adenosine residues10.70.05of some mRNAs.DUSP14Involved in the inactivation of MAP kinases.10.40.02Dephosphorylates ERK, JNK and p38 MAP-kinasesSLC27A6AMP-dependent synthetase and ligase10.30.05PTPRCAPProtein tyrosine phosphatase receptor type C-associated10.20.05protein; CD45-associated protein; Lymphocytephosphatase-associated phosphoproteinOSBPL7Oxysterol binding protein-related protein 7; OSBP-10.20.01related protein 7; ORP-7ZNF251ATP/GTP-binding site motif A (P-loop); KRAB box;10.20.05Zn-finger, C2H2 subtype;UBE2NThe UBE2V2/UBE2N heterodimer catalyzes the9.70.01synthesis of non-canonical poly-ubiquitin chains that arelinked through Lys-63. Mediates transcriptionalactivation of target genes. Plays a role in the control ofprogress through the cell cycle and differentiation.TBCATubulin-folding protein; involved in the early step of the9.60.04tubulin folding pathwayQ96NS3low complexity; transmembrane9.60.05KHKKetohexokinase; Hepatic fructokinase9.50.04GSTZ1Bifunctional enzyme showing minimal glutathione-9.40.04conjugating activity and low glutathione peroxidaseactivityANKRD5Ankyrin repeat domain protein 59.30.04UBXD2UBX domain-containing protein 29.30.04Q8N336;Protein of unknown function DUF6099.20.05ALOX5Arachidonate 5-lipoxygenase; 5-lipoxygenase; 5-LO9.20.05GPR174Putative receptor for purines coupled to G-proteins9.10.05TRIP4Transcription coactivator of nuclear receptors which9.00.05functions in conjunction with CBP-p300 and SRC-1 andmay play an important role in establishing distinctcoactivator complexes under different cellularconditions. Plays a pivotal role in the transactivation ofNF-kappa-B, SRF and AP1. Acts as a mediator oftransrepression between nuclear receptor and AP1 orNFκ-B.Q9P1G1signal peptide8.70.02Q96E44Beta and gamma crystallin; Nuclear protein SET8.50.02HCG9; HLA-JImmunoglobulin-like; Immunoglobulin/major8.50.05histocompatibility complex; Major histocompatibilitycomplex protein, class IQ9BUJ0Alpha/beta hydrolase; Esterase/lipase/thioesterase, active8.20.04sitePRLRCytokine receptor, common beta/gamma chain;8.00.02Fibronectin, type III; Long hematopoietin receptor,single chainDACH2Bipartite nuclear localization signal; Transforming8.00.03protein SkiO60384Zn-finger, C2H2 type7.90.03Q9BYE9Cadherin7.90.05TOP3BPossesses negatively supercoiled DNA relaxing activity7.40.06MPP4May play a role in retinal photoreceptors development7.30.05LNXE3 Ubiquitin ligase protein that mediates ubiquitination7.30.04and subsequent proteasomal degradation of NUMB.CYB5M;Cytochrome b5 is a membrane bound hemoprotein which7.20.05OMB5function as an electron carrier for several membranebound oxygenasesGRTP1RabGAP/TBC domain; Somatotropin hormone7.10.03Q9NZ13Zn-finger, C2H2 type6.90.03PROZAppears to assist hemostasis by binding thrombin and6.90.05promoting its association with phospholipid vesiclesHELIC1ATP/GTP-binding site motif A (P-loop); DEAD/DEAH6.80.04box helicase; Helicase, C-terminal; Sec63 domainEDG7Receptor for lysophosphatidic acid (LPA), a mediator of6.60.04diverse cellular activities. May play a role in thedevelopment of ovarian cancer. Seems to be coupled tothe G(i)/G(0) and G(q) families of heteromeric GproteinsEPM2ALikely multifunctional endocytic receptor that may be6.60.04implicated in the uptake of lipoproteins and of proteases.Binds LDL and receptor-associated protein (RAP).Could play a role in cell-cell interactionQ9UPS4coiled-coil; low complexity6.50.05Q96LQ8Alpha-2-macroglobulin; Alpha-2-macroglobulin, N-6.50.04terminalMYL4Regulatory light chain of myosin. Does not bind calcium6.50.04ACTL6Required for maximal ATPase activity of BRG1 and are6.50.03also required with BRG1 for association of the complexwith chromatin/matrixCHCHD5Bipartite nuclear localization signal6.40.03MFAP1Component of the elastin-associated microfibrils6.40.05PLAC8Placenta-specific gene 8 protein; C15 protein6.40.01LAMA3Laminin, binds to cells via a high affinity receptor is6.20.04thought to mediate the attachment, migration andorganization of cells into tissues during embryonicdevelopment by interacting with other extracellularmatrix componentsEDN1Endothelins are endothelium-derived vasoconstrictor6.20.05peptidesMSX1Acts as a transcriptional repressor. May play a role in6.10.04limb-pattern formation. Acts in cranofacial developmentand specifically in odontogenesisPKP4May play a role in junctional plaques6.10.05PRYTestis-specific PTP-BL related Y protein; PTPN13-like6.10.05proteinQ13862DNA-binding protein.6.00.04PAK2The activated kinase acts on a variety of targets.6.00.03Phosphorylates ribosomal protein S6, histone H4 andmyelin basic proteinWdr68WD-repeat protein 68; WD-repeat protein An115.90.02homologSENP6Protease that releases SUMO-1 from its precursor5.90.03sequenceDNMT2Its strong binding to DNA suggests that it may mark5.90.04specific sequences in the genome by binding to DNAthrough the specific target-recognizing motif. Not activeas a DNA methyltransferase.IL8IL-8 is a chemotactic factor that attracts neutrophils,5.70.01basophils, and T-cells, but not monocytes. It is alsoinvolved in neutrophil activation. It is released fromseveral cell types in response to an inflammatorystimulus.CCSDelivers copper to copper zinc superoxide dismutase5.60.02(SOD1)RGS14Inhibits signal transduction by increasing the GTPase5.60.05activity of G protein-α subunits resulting in their inactiveGDP-bound formARHGAP20RA domain; RhoGAP domain5.60.05COLEC10C-type lectin; Collagen triple helix repeat5.60.02Q96PN6ATP/GTP-binding site motif A (P-loop); Guanylate5.20.05cyclaseFBXW5Cyclin-like F-box; G-protein beta WD-40 repeat5.10.04ASTN2Fibronectin, type III5.10.03PCYT1BControls phosphatidylcholine synthesis5.00.06MUC11Actin-binding, actinin-type; Eukaryotic RNA polymerase4.90.04II heptapeptide repeatQ8NC34Immunoglobulin-like4.90.06Q9UFY9ATP/GTP-binding site motif A (P-loop); Cell4.90.03division/GTP binding proteinAPLP1May play a role in postsynaptic function. The C-terminal4.80.05gamma-secretase processed fragment, ALID1, activatestranscription activation through APBB1 (Fe65) binding.May interact with cellular G-protein signaling pathways.Q96N54Olfactory receptor; Rhodopsin-like GPCR superfamily4.80.02O60290KRAB box4.80.05COL5A1Collagen triple helix repeat; Fibrillar collagen, C-4.80.02terminal; Laminin G; Prenyl group binding site (CAAXbox)NRG1Direct ligand for ERBB3 and ERBB4 tyrosine kinase4.70.05receptors. Concomitantly recruits coreceptors, resultingin ligand-stimulated tyrosine phosphorylation andactivation of the ERBB receptors. Multiple isoformsperform diverse functions such as inducing growth anddifferentiation of epithelial, glial, neuronal, and skeletalmuscle cells.Q96FB5CGI-41 protein4.60.02CYP2J2This enzyme metabolizes arachidonic acid4.60.06predominantly via a NADPH-dependent olefinepoxidation to all four regioisomeric cis-epoxyeicosatrienoic acids.DBIBinds medium-and long-chain acyl-CoA esters with4.60.02very high affinity and may function as an intracellularcarrier of acyl-CoA esters. This protein may also act as aneuropeptide to modulate the action of the GABAreceptorHOOK1Cytoskeletal linker protein, which may be involved in4.60.04tethering membrane bound organelles to thecytoskeleton.FTCDFolate-dependent enzyme, that displays both transferase4.60.03and deaminase activity. Serves to channel one-carbonunits from formiminoglutamate to the folate poolCD84Leukocyte antigen CD84.4.50.04HIPK2Protein kinase acting as a corepressor of several4.50.02transcription factors, including SMAD1 andPOU4F1/Brn3a and probably NK homeodomaintranscription factors: Inhibits cell growth and promotesapoptosis. In response to TGFB, cooperates with DAXXto activate JNK. Phosphorylates the antiapoptotic factorCTBP1 and promotes its proteasomal degradation. In theWnt/beta-catenin signaling pathway acts as anintermediate kinase between TAK1 and NLK to promotethe proteasomal degradation of c-MybQ8TDS9putative G-protein coupled receptor GPCR42.4.40.05Q9H7B7low complexity; signal peptide4.40.05CCL20Chemotactic factor that attracts lymphocytes and,4.40.01slightly, neutrophils, but not monocytes. Inhibitsproliferation of myeloid progenitors in colony formationassays.NFKB2NFκB subunit; p52 and p100 are respectively the minor4.30.02and major form. Appears to have dual functions such ascytoplasmic retention of attached NFκB proteins andgeneration of p52 by a cotranslational proteasome-mediated processing. p52 binds to the κB consensussequence “GGRNNYYCC-3”, located in the enhancerregion of genes involved in immune response and acutephase reactions.C13orf11coiled-coil; low complexity; signal peptide;4.30.06transmembraneMKKSMay play a role in protein processing in limb, cardiac4.30.06and reproductive system developmentSLC6A1Terminates the action of GABA by its high affinity4.20.02sodium-dependent reuptake into presynaptic terminalsBAZ2AMay play a role in transcriptional regulation interacting4.20.01with ISWI. May serve a specific role in maintaining oraltering the chromatin structure of the rDNA locusSLC16A10T-type amino acid transporter 1; solute carrier family 16,4.20.05# 10Q8NG48WINS1 protein isoform 1.4.20.05ARHGEF1Seems to play a role in the regulation of RhoA GTPase4.20.00by guanine nucleotide-binding α-12 andα-13 subunits.GTPase-activating protein (GAP) for these subunits, andas guanine nucleotide exchange factor (GEF) for RhoAGTPase; stimulates the RhoGEF activity.COL9A2Structural component of hyaline cartilage and vitreous of4.10.05the eyeCTLA4Possibly involved in T-cell activation. Binds to B7-14.10.02(CD80) and B7-2 (CD86)SYNGR2Synaptogyrin-2; Cellugyrin4.10.05PSMA1Proteasome, a multicatalytic proteinase complex with an4.10.04ATP-dependent proteolytic activityEPN1Binds to membranes enriched in phosphatidylinositol-4.10.054,5-biphosphate. Modifies membrane curvature,facilitates the formation of clathrin-coated invaginations,regulates receptor-mediated endocytosisTMSB10Important role in the organization of the cytoskeleton.4.00.06Binds to and sequesters actin monomers (G actin) andtherefore inhibits actin polymerizationTCN1Vitamin B12-binding protein. Transports cobalamin into4.00.03cellsRBM3Putative RNA-binding protein 3; RNA-binding motif4.00.01protein 3C20orf26Protein C20orf264.00.06HYAL4EGF-like domain; Glycoside hydrolase, family 56;3.80.05Glycoside hydrolase, family 56, sperm surface proteinPH20; Multicopper oxidase, type 1BACH1Transcriptional regulator that acts as repressor or3.80.04activator. Binds, in-vitro, to NF-E2 binding sites. Playimportant roles in coordinating transcription activationand repression by MAFKPMAIP1Phorbol-12-myristate-13-acetate-induced protein 1;3.80.02Q96T82signal peptide; transmembrane3.70.00TRACT-cell receptor alpha chain C region3.70.04KLF8Transcriptional repressor. Binds to CACCC-box3.70.04promoter elementsDVL2May play a role in the signal transduction pathway3.70.02mediated by multiple Wnt genesCRK7Cell division cycle 2-related protein kinase 73.70.04Q9H631Mak10 subunit, NatC N(alpha)-terminal3.60.05acetyltransferaseZNF208KRAB box; Neutral zinc metallopeptidases; Zn-finger,3.60.05C2H2 subtype;MAP2K4Dual specificity kinase that activates the JUN kinases3.60.05MAPK8 (JNK1) and MAPK9 (JNK2) as well asMAPK14 (p38) but not MAPK1 (ERK2) or MAPK3(ERK1)CENTB2GTPase-activating protein for the ADP ribosylation3.60.03factor familyQ8N958unknown3.60.03Q9BYA6low complexity3.50.04ZFYVE20Zn-finger, C2H2 type; Zn-finger, FYVE type3.50.02DSTNActin-depolymerizing protein. Severs F-actin filaments3.50.02and binds to actin monomers (G-actin), in pH-independent mannerITGB4Integrin alpha-6/beta-4 is a receptor for laminin. It plays3.50.03a critical structural role in the hemidesmosome ofepithelial cellsM6PRTransport of phosphorylated lysosomal enzymes from3.50.05the Golgi complex and the cell surface to lysosomes.NEF3Neurofilaments usually contain 3 intermediate filament3.40.01proteins: L, M, and H involved in maintenance ofneuronal caliberQ8N4T82,3-dihydro-2,3-dihydroxybenzoate dehydrogenase;3.40.03Glucose/ribitol dehydrogenase;ZNF36May be involved in transcriptional regulation3.40.02CCL2Chemotactic factor that attracts monocytes and basophils3.40.02but not neutrophils or eosinophils. Has been implicatedin the pathogenesis of diseases characterized bymonocytic infiltrates, like psoriasis, rheumatoid arthritisor atherosclerosis. May be involved in the recruitment ofmonocytes into the arterial wall during the diseaseprocess of atherosclerosisANKMY1Ankyrin repeat and MYND domain protein 1; Testis-3.40.05specific ankyrin-like protein 1; Zinc-finger MYNDdomain protein 13ABCC13Putative ATP-binding cassette transporter C133.40.04Q86TW0Bipartite nuclear localization signal; Zn-finger, C-x8-C-3.40.03x5-C-x3-H typeZNF213May be involved in transcriptional regulation3.40.04HNF4ATranscriptionally controlled transcription factor. Binds to3.30.01DNA sites required for the transcription of alpha 1-antitrypsin, apolipoprotein CIII, transthyretin genes andHNF1-alpha.MAFFInteracts with the upstream promoter region of the3.30.01oxytocin receptor gene. May be a transcriptionalenhancer. May also serve as transcriptional activator bydimerizing with other basic-zipper proteins andrecruiting them to specific DNA-binding sites. May beinvolved in the cellular stress responseFBXO32Probably recognizes and binds to some phosphorylated3.30.03proteins and promotes their ubiquitination anddegradation during skeletal muscle atrophyQ9P233Calponin-like actin-binding; Eggshell protein; Leucine-3.30.05rich repeatTNFPro-inflammatory cytokine tumour necrosis factor α that3.30.02binds to TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR.CXCL2Hematoregulatory chemokine, which, in vitro, suppresses3.30.00hematopoietic progenitor cell proliferation. Produced byactivated monocytes & neutrophils and expressed at sitesof inflammation.Q8N233NHL repeat; Zn-finger, C2H2 type3.30.03MAKCould play an important function in spermatogenesis3.30.03GPR30Orphan receptor; possibly for a chemokine3.20.01MAGMASMitochondria-associated granulocyte macrophage CSF3.20.05signaling molecule, mitochondrial precursorLHX2Transcriptional regulatory protein involved in the control3.20.05of cell differentiation in developing lymphoid and neuralcell typesQ96LP3Leucine-rich repeat3.20.05Q9NXD2Bipartite nuclear localization signal3.20.02FOXQ1Forkhead box protein Q1; Hepatocyte nuclear factor 33.20.01forkhead homolog 1;TFECBasic helix-loop-helix dimerization domain bHLH3.10.03Q96EC8Protein of unknown function DUF6493.10.03PTGS2May have a role as a major mediator of inflammation3.10.01and/or a role for prostanoid signaling in activity-dependent plasticityQ8N3K5Cysteine-rich flanking region, N-terminal;3.10.02Immunoglobulin-like; Leucine-rich repeat; RNA-bindingregion RNP-1DNAH5ATP/GTP-binding site motif A (P-loop); Dynein heavy3.00.01chain; Eukaryotic thiol (cysteine) proteaseHDCHistidine decarboxylase; HDC3.00.00C6orf149Bipartite nuclear localization signal; Complex 1 LYR3.00.06proteinPTGISCatalyzes the isomerization of prostaglandin H2 to3.00.06prostacyclinIL6IL-6 is a cytokine with a wide variety of biological2.90.00functions: it plays an essential role in the finaldifferentiation of B-cells into Ig-secreting cells, itinduces myeloma and plasmacytoma growth, it inducesnerve cells differentiation, in hepatocytes it induces acutephase reactantsCXCL1Has chemotactic activity for neutrophils. May play a role2.90.01in inflammation and exerts its effects on endothelial cellsin an autocrine fashion.KPNB1Functions in nuclear protein import, either in association2.90.05with an adapter protein, like an importin-alpha subunit,which binds to nuclear localization signals (NLS) incargo substrates, or by acting as autonomous nucleartransport receptor.CD81May play an important role in the regulation of2.80.05lymphoma cell growth. May acts as the viral receptor forHCVGNSN-acetylglucosamine-6-sulfatase precursor;2.80.03PTSInvolved in the biosynthesis of tetrahydrobiopterin, an2.80.01essential cofactor of aromatic amino acid hydroxylases.MYLIPBand 4.1 domain; Ezrin/radixin/moesin ERM2.80.01Q96IT8low complexity2.80.02SLC17A4Na/PO4 cotransporter; solute carrier family 17, member 42.80.02NPAS2Neuronal PAS domain protein 2; MOP42.80.00PANK3Plays a role in the physiological regulation of the2.80.00intracellular CoA concentrationPHLDA2Pleckstrin-like2.80.02Q96MX1down-regulated by Ctnnb1, a.2.80.06SP3Binds to GT and GC boxes promoters elements. Probable2.80.06transcriptional activatorQ9Y3U6low complexity2.70.01NRP1The membrane-bound isoform 1 is a receptor involved in2.70.04the development of the cardiovascular system, in VEGF-induced angiogenesis, in the formation of certainneuronal circuits and in organogenesis outside thenervous system.NCK1Adapter protein which associates with tyrosine-2.70.03phosphorylated growth factor receptors or their cellularsubstratesQ8WUC7Bipartite nuclear localization signal2.70.02Q9P2X3Protein of unknown function UPF00292.70.01NICE1NICE-1 protein2.70.06SH3YL1Protein of unknown function DUF500; SH3 domain2.70.01CCL23Shows chemotactic activity for monocytes, resting T-2.70.03lymphocytes, and neutrophils, but not for activatedlymphocytes.SNAPAPMay modulate a step between vesicle priming, fusion2.70.05and calcium-dependent neurotransmitter release. Itsphosphorylation state influences exocytotic proteininteractions and may regulate synaptic vesicleexocytosis. May also have role in SNARE-mediatedmembrane fusion in non-neuronal cellsZHX2Homeobox; Zn-finger, C2H2 type2.60.03LOXL2Lysyl oxidase homolog 2 precursor; Lysyl oxidase-2.60.01related protein WS9-14ACSL6Activation of long-chain fatty acids for both synthesis of2.60.05cellular lipids, and degradation via beta-oxidation. Playsan important role in fatty acid metabolism in brainQ96BW9unknown2.60.05Q9BT00RPA interacting protein2.60.03Q9UJA5Bipartite nuclear localization signal; Eukaryotic initiation2.60.03factor 3, gamma subunitICAM3ICAM proteins are ligands for the leukocyte adhesion2.60.04LFA-1 protein (integrin alpha-L/beta-2). ICAM3 is also aligand for integrin alpha-D/beta-2TENC1Bipartite nuclear localization signal; Protein kinase C,2.60.01phorbol ester/diacylglycerol binding; SH2 motifNME1Major role in the synthesis of nucleoside triphosphates2.50.02other than ATPIL1F9Function as an agonist of NFκB activation through the2.50.04orphan IL-1-receptor-related protein 2. Could constitutepart of an independent signaling system analogous tointerleukin-1α, and β receptor agonist and interleukin-1receptor type I (IL-1R1), that is present in epithelialbarriers and takes part in local inflammatory responseQ8WVV9RNA-binding region RNP-1; Serine/threonine2.50.05dehydratase, pyridoxal-phosphate attachment siteZNF177May be involved in transcriptional regulation2.50.02DNASE1Seems to be involved in cell death by apoptosis. Binds2.50.01specifically to G-actin and blocks actin polymerizationQ8N867unknown2.50.04SOD2Destroys radicals which are normally produced within2.50.01the cells and which are toxic to biological systemsQ9H9C7BRCT domain2.50.04C16orf3Protein C16orf32.50.02Q9BRJ9Basic helix-loop-helix dimerization domain bHLH2.50.04GDACatalyzes the hydrolytic deamination of guanine,2.50.01producing xanthine and ammoniaLILRB1Receptor for class I MHC antigens. Recognizes a broad2.50.05spectrum of HLA-A, HLA-B, HLA-C and HLA-Galleles. Ligand binding results in inhibitory signals anddown-regulation of the immune response. Engagement ofLILRB1 present on NK cells or T-cells by class I MHCmolecules protects the target cells from lysis.HBG1The epsilon chain is a beta-type chain of early2.50.04mammalian embryonic hemoglobinMAP3K8Able to activate NFκB 1 by stimulating proteasome-2.50.06mediated proteolysis of NFκB 1/p105. Plays a role in thecell cycle.Q8WVI0low complexity2.50.02HELLSBipartite nuclear localization signal; Helicase, C-2.50.02terminal; SNF2 related domainCDK11Protein kinase; Serine/Threonine protein kinase2.40.05ARHGDIARegulates the GDP/GTP exchange reaction of the Rho2.40.02proteins by inhibiting GDP dissociation and subsequentGTP bindingQ9H759Immunoglobulin-like2.40.04SERPINB8Inhibits urokinase-type plasminogen activator. The2.40.04monocyte derived PAI-2 is distinct from the endothelialcell-derived PAI-1Q86VU9Proline-rich region2.40.00Q8IUZ5Aminotransferase class-III2.40.05Q9BWJ2unknown2.40.05ARG2May play a role in the regulation of extra-urea cycle2.40.04arginine metabolism and also in down-regulation ofnitric oxide synthesis.TNFAIP6Possibly involved in cell-cell and cell-matrix interactions2.40.01during inflammation and tumorigenesisC14orf132transmembrane2.40.05RIPK2Activates pro-caspase-1 and pro-caspase-8. Potentiates2.40.01CASP-8-mediated apoptosis. Activates NF-kappa-BSLC39A8Zinc transporter ZIP2.40.02ATP2B1This magnesium-dependent enzyme catalyzes the2.30.01hydrolysis of ATP coupled with the transport of calciumout of the cellCRKLMay mediate the transduction of intracellular signals2.30.02PABPC3Binds the poly(A) tail of mRNA. May be involved in2.30.05cytoplasmic regulatory processes of mRNA metabolism.ERCC6Is involved in the preferential repair of active genes.2.30.06Presumed DNA or RNA unwinding function.Q8NC30transmembrane2.30.00Q969W3low complexity2.30.00HTR1FOne of the several different receptors for serotonin, a2.30.01biogenic hormone that functions as a neurotransmitter, ahormone, and a mitogen. Activity mediated by Gproteins that inhibit adenylate cyclase activityJARID1DMay play a role in spermatogenesis2.30.01Q9HBM0Plays a pivotal role in the establisment of adherens2.30.06junctions and their maintenance in adult lifeNOVImmediate-early protein likely to play a role in cell2.30.04growth regulationKIAA1533GRAM domain2.30.00VPREB1Associates with the Ig-mu chain to form a molecular2.30.00complex that is expressed on the surface of pre-B-cellsand presumably regulates Ig gene rearrangements inearly B-cell differentiationQ96NJ4signal peptide2.30.02FCGR2BLow affinity receptor for the Fc region of complexed2.30.03immunoglobulins gamma. receptor. Involved in variouseffector and regulatory functions such as phagocytosis ofimmune complexes and modulation of antibodyproduction by B-cells;ENSG00000110900CD9/CD37/CD63 antigen2.30.00Q9HCK1low complexity2.30.06BCDO2Asymmetrically cleaves beta-carotene at the double2.30.04bond resulting in the formation of beta-carotenal andbeta-ionone. Lycopene is also oxidatively cleaved.HOXB2Sequence-specific transcription factor which is part of a2.30.03developmental regulatory system that provides cells withspecific positional identities on the anterior-posterior axisTUSC4Bipartite nuclear localization signal2.30.03Q8NBF0low complexity; signal peptide; transmembrane2.30.03ABL2Tyrosine-protein kinase ABL2; Tyrosine kinase ARG2.30.02Q8TF23BED finger; Cytochrome c heme-binding site; KRAB2.30.02box; Zn-finger, C2H2 subtypeQ8IUC6Proline-rich extensin2.30.03CD151Essential for the proper assembly of the glomerular and2.30.00tubular basement membranes in kidneyIER3Immediate early response 3 protein; Radiation-inducible2.30.03immediate-early gene IEX-1; GLY96; PACAP-responsive Differentiation-dependent gene 2 protein;DIF-2 proteinQ9NXL6SID1 transmembrane family, member 12.30.06C14orf79unknown2.30.03C16orf44BTB/POZ domain; Kelch repeat2.30.02KIAA1404Protein KIAA14042.30.00ENSG00000106603signal peptide; transmembrane2.20.05Q86XN7;Aldehyde dehydrogenase; Proline-rich extensin2.20.02Q9H9M1RAB3BProtein transport. Probably involved in vesicular traffic2.20.04RHOCProtein phosphatase 2C-like2.20.04NDUFB4Transfer of electrons from NADH to the respiratory2.20.02chain. The immediate electron acceptor is believed to beubiquinoneSTAMBPBipartite nuclear localization signal; Mov34 family2.20.03ECE2Converts big endothelin-1 to endothelin-12.20.05LCP2Involved in T cell antigen receptor mediated signaling2.20.04OASLBinds double-stranded RNA and DNA, but no OAS2.20.03activityCEBPDC/EBP is a DNA-binding protein that recognizes two2.20.02different motifs: the CCAAT homology common tomany promoters and the enhanced core homologycommon to many enhancers. Important transcriptionalactivator in the regulation of genes involved in immuneand inflammatory responses, may play an important rolein the regulation of the several genes associated withactivation and/or differentiation of macrophagesSOD2Destroys radicals which are normally produced within2.20.02the cells and which are toxic to biological systemsQ8N316Protein kinase; Serine/Threonine protein kinase2.20.01KIAA1847Zinc finger CCCH-type with G patch domain protein 92.10.00Q9UFQ7low complexity; transmembrane2.10.04ADAMTS6ADAMTS-6 precursor; A disintegrin and2.10.02metalloproteinase with thrombospondin motifs 6S100A12Calcitermin possesses antifungal activity against2.10.04C. albicans & is also active vs. E. coli, P. aeruginosa butnot Listeria and S. aureusDBC1Deleted in bladder cancer chromosome region candidate2.10.051.KRTAP2-4Keratin, high sulfur B2 protein; von Willebrand factor,2.10.00type CPEMTCatalyzes three sequential methylation of PE by AdoMet,2.10.05thus producing phosphatidylcholineTNIP1Interacts with zinc finger protein A20/TNFAIP3 and2.10.05inhibits TNF-induced NFκB-dependent gene expressionby interfering with an RIP- or TRAF2-mediatedtransactivation signal. Over-expression can inhibit HIVreplication.PTP4A3Prenyl group binding site (CAAX box); Tyrosine2.10.00specific protein phosphatase and dual specificity proteinphosphataseSOD3Destroys radicals which are normally produced within2.10.04the cells and which are toxic to biological systemsADORA2AReceptor for adenosine. The activity of this receptor is2.10.05mediated by G proteins which activate adenylyl cyclaseC14orf166Protein C14orf1662.10.06PELI1Scaffold protein involved in the IL-1 signaling pathway2.10.01via its interaction with the complex containing IRAKkinases and TRAF6. Required for NFκB activation andIL-8 gene expression in response to IL-1PPP1R15Bprotein phosphatase 1, regulatory subunit 15B.2.10.05KLHL18;Kelch-like protein 182.10.02CHCHD3Protein of unknown function DUF7372.10.04O94940SAM (and some other nucleotide) binding motif2.10.01TEX15testis expressed sequence 15.2.10.05DAB1Adapter molecule functioning in neural development.2.10.05May regulate SIAH1 activityRAB5BProtein transport. Probably involved in vesicular traffic2.10.02IGHG3Ig alpha is the major immunoglobulin class in body2.10.06secretions. It may serve both to defend against localinfection and to prevent access of foreign antigens to thegeneral immunologic systemINSIG1May play a role in growth and differentiation of tissues2.10.03involved in metabolic control. May play a regulatory roleduring G0/G1 transition of cell growthCXCL6Chemotactic for neutrophil granulocytes2.10.03GPR84Rhodopsin-like GPCR superfamily2.10.04Q96G36Alpha tubulin; Epsilon tubulin; Tubulin/FtsZ protein2.00.01Q8NFQ8AF464140.2.00.03Q9BRC8low complexity; transmembrane2.00.03CACNA1HVoltage-sensitive calcium channels (VSCC) mediate the2.00.00entry of calcium ions into excitable cells and are alsoinvolved in a variety of calcium-dependent processes,including muscle contraction, hormone orneurotransmitter release, gene expression, cell motility,cell division and cell death.ABHD5Alpha/beta hydrolase; Esterase/lipase/thioesterase, active2.00.01site; Prolyl aminopeptidase S33Q8NBR8Immunoglobulin-like; Ig/major histocompatibility2.00.02complexPOLR2DDNA-dependent RNA polymerase catalyzes the2.00.06transcription of DNA into RNA using the fourribonucleoside triphosphates as substrates. Associateswith POLR2GO76052low complexity2.00.05Q8N7I3Immunoglobulin-like2.00.02Q9Y3B9Bipartite nuclear localization signal2.00.06HIF1AFunctions as a master transcriptional regulator of the2.00.02adaptive response to hypoxia. Under hypoxic conditionsactivates the transcription of over 40 genes, including,erythropoietin, glucose transporters, glycolytic enzymes,vascular endothelial growth factor, and other geneswhose protein products increase oxygen delivery orfacilitate metabolic adaptation to hypoxia. Plays anessential role in embryonic vascularization, tumorangiogenesis and pathophysiology of ischemic disease.Q9BTK5G-protein beta WD-40 repeat2.00.04PHKG1Phosphorylase b kinase catalyzes the phosphorylation of2.00.02serine in certain substrates, including troponin IPLEKHB2Pleckstrin-like; Proline-rich extensin2.00.02CCNB1Essential for the control of the cell cycle at the G2/M2.00.00(mitosis) transitionO15069Nascent polypeptide-associated complex NAC2.00.05EIF5APrecise role of eIF-5A in protein biosynthesis is not2.00.05known but it functions by promoting the formation of thefirst peptide bondHDAC10Responsible for the deacetylation of lysine residues on2.00.01the N-terminal part of the core histones (H2A, H2B, H3and H4). Histone deacetylation gives a tag for epigeneticrepression and plays an important role in transcriptionalregulation, cell cycle progression and developmentalevents.Q9H908unknown2.00.05BTBD12BTB/POZ domain2.00.05RBMS3Paraneoplastic encephalomyelitis antigen; RNA-binding1.90.04region RNP-1FBXL12Substrate-recognition component of the SCF (SKP1-1.90.04CUL1-F-box protein)-type E3 ubiquitin ligase complexSPG72Fe—2S ferredoxin; Peptidase M411.90.01MGAT5Catalyzes the addition of N-acetylglucosamine in beta 1-1.90.026 linkage to the alpha-linked mannose of biantennary N-linked oligosaccharides.SIAT4CIt may catalyze the formation of the NeuAc-alpha-2,3-1.90.01Gal-beta-1,3-GalNAc- or NeuAc-alpha-2,3-Gal-beta-1,3-GlcNAc-sequences found in terminal carbohydrategroups of glycoproteins and glycolipids.IL12BCytokine that can act as a growth factor for activated T1.90.00and NK cells, enhances the lytic activity ofNK/lymphokine-activated killer cells, stimulates theproduction of IFN-γ by resting PBMCGGNProline-rich extensin; Proline-rich region1.90.01NFKBIAInhibits NFκB by complexing with and trapping it in the1.90.00cytoplasm. May be involved in regulation oftranscriptional responses to NF-kappa-B, including celladhesion, immune and proinflammatory responses,apoptosis, differentiation and growth.ENSG00000153820low complexity1.90.01CYP19A1Catalyzes the formation of aromatic C18 estrogens from1.90.01C19 androgensMUC13EGF-like domain; SEA domain1.90.02CGI-117Protein CGI-1171.90.00HRMT1L6Mono and asymmetric dimethylation of guanidino1.90.02nitrogens of arginyl residues present in a glycine andarginine-rich domainWTAPWilms-tumor 1-associating protein; Putative pre-mRNA1.90.05splicing regulator female-lethal(2D) homologDPYSDihydropyrimidinase; DHPase; Hydantoinase; DHP1.90.05LILRB2Receptor for class I MHC antigens. Recognizes a broad1.90.06spectrum of HLA-A, HLA-B, HLA-C and HLA-Galleles. Involved in the down-regulation of the immuneresponse and the development of tolerance.HIAT1General substrate transporter; Sugar transporter1.90.04superfamily; Tetracycline resistance proteinKDELR1Required for the retention of luminal endoplasmic1.90.04reticulum proteins. Required for normal vesicular trafficthrough Golgi.GPC5Cell surface proteoglycan that bears heparan sulfate1.90.05ATP4ACatalyzes the hydrolysis of ATP coupled with the1.90.05exchange of H(+) and K(+) ions across the plasmamembrane. Responsible for acid production in thestomachC5orf13Neuronal protein 3.1; p311 protein1.90.05KLK14Kallikrein-14 precursor; Kallikrein-like protein 6; KLK-1.90.00L6Q9NZY8unknown1.90.02ZNF80May be involved in transcriptional regulation1.90.04SMARCA2Transcriptional coactivator cooperating with nuclear1.90.02hormone receptors to potentiate transcriptional activationMXD3Basic helix-loop-helix dimerization domain bHLH1.90.03PDCD6Calcium-binding protein required for T cell receptor-,1.90.02Fas-, and glucocorticoid-induced cell death. May mediateCa(2+)-regulated signals along the death pathwayCYorf14Hypothetical protein CYorf141.90.02C20orf97Disrupts insulin signaling by binding directly to Akt1.90.05kinases and blocking their activation. Interacts withMAPK kinases and regulates activation of MAP kinases.Q9HCM3Bacterial regulatory protein, LuxR family1.90.03PFKFB3Synthesis and degradation of fructose 2,6-bisphosphate1.90.02NID2Cell adhesion glycoprotein which is widely distributed in1.90.05base-ment membranes. Binds to collagens I & IV,perlecan, laminin 1. Probably has role in cell-extracellular matrix interactionsQ8IZ83Aldehyde dehydrogenase1.90.03Q9Y3H6Bipartite nuclear localization signal1.90.04FCN3Involved in the serum exerting lectin activity.1.90.01Q9NVS3IQ calmodulin-binding region1.80.02SEH1LNucleoporin SEH1-like; SEC13-like protein1.80.01C6orf115Protein C6orf1151.80.04LRP16Protein LRP161.80.02TFRCTransferrin receptor is necessary for development of1.80.05erythrocytes and the nervous system. Cellular uptake ofiron occurs via receptor-mediated endocytosis of ligand-occupied transferrin receptor into specialized endosomes.MAP1LC3AProbably involved in formation of autophagosomal1.80.05vacuolesQ7Z4E7low complexity1.80.01O60739Probably involved in translation1.80.00LGR7Receptor for relaxins. The activity of this receptor is1.80.05mediated by G proteins leading to stimulation ofadenylate cyclase and an increase of cAMP.CFHL5Involved in complement regulation1.80.01Q8N9G6low complexity1.80.05ICAM1ICAM proteins are ligands for the leukocyte adhesion1.80.01LFA-1 protein (Integrin alpha-L/beta-2)Q9H606Proline-rich region1.80.06ENSG0000154511low complexity; transmembrane1.80.06SLC24A2Critical component of the visual transduction cascade.1.80.03Q9H8S7Bipartite nuclear localization signal1.80.00TRAPPC4May play a role in vesicular transport from endoplasmic1.80.03reticulum to GolgiZNF513Zn-finger, C2H2 type1.80.03CLK3Phosphorylates seine- and arginine-rich (SR) proteins of1.80.01the spliceosomal complex may be a constituent of anetwork of regulatory mechanisms that enable SRproteins to control RNA splicing. Phosphorylates serines,threonines and tyrosinesQ9NTF2Prenyl group binding site (CAAX box)1.80.04SIGLEC10Putative adhesion molecule that mediates sialic-acid1.80.02dependent binding to cells.SLC22A11General substrate transporter1.80.03AQP9Forms a channel with a broad specificity. Mediates1.80.02passage of a wide variety of non-charged solutes.SSBP3May be involved in transcription regulation of the alpha1.80.052(I) collagen gene where it binds to the single-strandedpolypyrimidine sequences in the promoter regionMAPK11Kinase involved in a signal transduction pathway that is1.80.05activated by changes in the osmolarity of theextracellular environment, by cytokines, or byenvironmental stress. Phosphorylates ATF2MAPK6Phosphorylates microtubule-associated protein 21.80.02(MAP2). May promote entry in the cell cycleGHRReceptor for pituitary gland growth hormone involved in1.80.03regulating postnatal body growth. On ligand binding,couples to the JAK2/STAT5 pathwayTREX1Exonuclease; Proline-rich region1.80.03CBARA1Bipartite nuclear localization signal; Calcium-binding1.80.01EF-handQ96RH9MUSP1.1.80.05MTHFD2Bifunctional NAD-dependent methylenetetrahydrofolate1.80.04dehydrogenase/cyclohydrolase, mitochondrial precursorPLXNA4Cell surface receptor IPT/TIG; Plexin1.80.01KIAA0084Hypothetical protein KIAA0084; HA20221.80.04RPS940S ribosomal protein S91.80.02PSMD1Acts as a regulatory subunit of the 26 proteasome which1.80.02is involved in ATP-dependent degradation ofubiquitinated proteinsQ9Y6U7Proline-rich region; Zn-finger, RING1.80.03RIMBP2Plays role in the synaptic transmission as bifunctional1.80.02linker.PAPMight be a stress protein involved in the control of1.80.05bacterial proliferationZNF274May function as a transcriptional repressor1.70.04ZIM2May function as a transcription factor1.70.05TFPIInhibits factor Xa directly and, in a Xa-dependent way,1.70.01inhibits VIIa/tissue factor activity, presumably byforming a quaternary Xa/LACI/VIIa/TF complex. Itpossesses an antithrombotic action and also the ability toassociate with lipoproteins in plasmaALOX15Converts arachidonic acid to 15S-1.70.04hydroperoxyeicosatetraenoic acid. Acts on C-12 ofarachidonate as well as on linoleic acidLAIR1Immunoglobulin-like1.70.04Q8N3D0low complexity1.70.03HYAL2Hyaluronidase that hydrolyzes high molecular weight1.70.03hyaluronic acid to produce an intermediate-sizedproduct.EHD1Acts in early endocytic membrane fusion and membrane1.70.01trafficking of recycling endosomesC1orf22Putative aipha-mannosidase C1orf221.70.01GPR52Orphan receptor1.70.03Q96CX6Leucine-rich repeat1.70.01Q9BYX4CARD interaction domain; DEAD/DEAH box helicase1.70.04SHOX2May be growth regulator and have a role in specifying1.70.05neural systems involved in processing somatosensoryinformation.PPP2R1AThe PR65 subunit of protein phosphatase 2A serves as a1.70.00scaffolding molecule to coordinate the assembly of thecatalytic subunit and a variable regulatory B subunitATRPhosphatidylinositol 3- and 4-kinase, FAT; FATC;1.70.04KIAA2010EVH1; Protein of unknown function DUF6251.70.01MPIInvolved in the synthesis of the GDP-mannose and1.70.03dolichol-phosphate-mannose required for criticalmannosyl transfersQ9H9X6Bipartite nuclear localization signal1.70.03SPIBSequence specific transcriptional activator which binds1.70.03to the PU-box, a purine-rich DNA sequence and can actas a lymphoid-specific enhancer. Promotes developmentof plasmacytoid dendritic cells (pDCs), also known astype 2 DC precursors (pre-DC2) or natural interferon(IFN)-producing cells. These cells have the capacity toproduce large amounts of interferon and block viralreplication.Q9BW08Bipartite nuclear localization signal; PWWP domain1.70.05EPS15Involved in cell growth regulation. May be involved in1.70.01the regulation of mitogenic signals and control of cellproliferation. Involved in the internalization of ligand-inducible receptors of the receptor tyrosine kinase (RTK)type, in particular EGFRDPH5Required for the methylation step in diphthamide1.70.04biosynthesisRELProto-oncogene that may play a role in differentiation1.70.01and lymphopoiesis. May function as a transcriptionaltransactivatorSIAT8AInvolved in the production of GD3 and GT3 from GM31.70.02ADAMTS13Neutral zinc metalloprotease ADAM/reprolysin M12B1.70.04LY86May cooperate with CD180 and TLR4 to mediate the1.70.01innate immune response to bacterial LPS and cytokineproduction. Important for efficient CD180 cell surfaceexpressionFBLN2Its binding to fibronectin and some other ligands is Ca1.70.03dependentADPRHL1ADP-ribosylglycohydrolase1.70.01TRPV6Ankyrin; Ion transport protein1.70.04TSC22D1Transcriptional repressor. Acts on the C-type natriuretic1.70.01peptide (CNP) promoterSASSarcoma amplified sequence; Tetraspanin-31; Tspan-311.70.01ASCL1Activates transcription by binding to the E box and may1.70.02play a role at early stages of development of specificneural lineages.Q9BVM2DPCD protein.1.70.01PRPF8Bipartite nuclear localization signal; Mov34 family1.70.06KPNA4Functions in nuclear protein import as an adapter protein1.70.01for nuclear receptor KPNB1. Binds specifically anddirectly to substrates containing either a simple orbipartite NLS motif.GLB1LGlycoside hydrolase, family 351.70.04CCNB1IP1E3 ubiquitin ligase. Modulates cyclin B levels and1.70.01participates in the regulation of cell cycle progressionthrough the G2 phase.CD44Receptor for hyaluronic acid (HA). Mediates cell-cell1.70.00and cell-matrix interactions through its affinity for HA.Also involved in lymphocyte activation, recirculationand homing, and in hematopoiesis.Q8NEQ3unknown1.70.03Q8N2I6;BTB/POZ domain; G-protein beta WD-40 repeat; K+1.70.03Q8TBC3channel tetramerisationSNAPC2Part of the SNAPc complex required for the transcription1.70.06of both RNA polymerase II and III small-nuclear RNAgenes.ARFRP1Possibly involved in plasma membrane-related signaling1.70.02eventsARTNProline-rich region; Transforming growth factor beta1.70.04(TGFb)LSM10Binds specifically to U7 snRNA1.70.00O60844Jacalin-related lectin1.70.05PDCD4Initiation factor eIF-4 gamma, MA31.70.05RIMS2Rab effector involved in exocytosis. May act as scaffold1.70.02proteinQ86W66esophageal cancer associated protein.1.70.01AP2S1Component of the adaptor complexes which link clathrin1.70.05to receptors in coated vesicles.Q8WUB2protein predicted by clone 23733.1.70.03GCH1Isoform GCH-1 is the functional enzyme, the potential1.70.02function of the euzymatically inactive isoforms remainsunknownQ9UPX5ATP/GTP-binding site motif A (P-loop)1.70.06NUDT4NUDIX hydrolase1.70.06HSPB1Involved in stress resistance and actin organization1.70.02Q9H679low complexity; signal peptide; transmembrane1.70.05ENSG0000087116Immunoglobulin-like1.70.01MAN2B1Necessary for the catabolism of N-linked carbohydrates1.70.02released during glycoprotein turnover.GKKey enzyme in the regulation of glycerol uptake and1.70.02metabolismNUTF2Facilitates protein transport into the nucleus. Interacts1.70.01with the nucleoporin p62 and with Ran.Q9H8H0coiled-coil; low complexity1.70.05GALNT9Ricin B lectin domain1.70.02F13A1Factor XIII is activated by thrombin and calcium ion to a1.60.05transglutaminase that catalyzes the formation of cross-links between fibrin chains, thus stabilizing the fibrinclot.Q96AP0low complexity1.60.05RPS940S ribosomal protein S91.60.02SLC2A14Facilitative glucose transporter. Probably a neuronal1.60.01glucose transporterATP5F1ATP synthase B chain, mitochondrial precursor1.60.04DDX21Can unwind double-stranded RNA (helicase) and can1.60.01fold or introduce a secondary structure to a single-stranded RNA (foldase). Functions as cofactor for c-Jun-activated transcription.TTLL3Tubulin tyrosine ligase-like protein 3; HOTTL1.60.06SMPDL3AAcid sphingomyelinase-like phosphodiesterase 3a1.60.03precursor;CPN2May play important roles in selective fasciculation and1.60.01zone-to-zone projection of the primary olfactory axonsAURKAIP1;May act as a negative regulator of Aurora-A kinase, by1.60.02AIP; AKIPdown-regulation through proteasome-dependentdegradationQ9P1V9low complexity1.60.02Q9NZE3KH domain, type 1; Zn-finger, RING1.60.03SLC29A1Delayed-early response protein/equilibrative nucleoside1.60.04transporterC1orf24Niban protein1.60.02Q9H3U1Armadillo repeat; TPR repeat1.60.01ARF4Involved in protein trafficking; may modulate vesicle1.60.03budding and uncoating within the Golgi apparatusZNF185May be involved in the regulation of cellular1.60.03proliferation and/or differentiationNUBP1Nucleotide-binding protein 1; NBP 11.60.04GPR25Orphan receptor1.60.01Q9Y2K2Protein kinase; Serine/Threonine protein kinase;1.60.01Tyrosine protein kinaseLILRA1May act as soluble receptor for class I MHC antigens1.60.05ENSG00000173961Bipartite nuclear localization signal; HMG1/2 (high1.60.05mobility group) box; High mobility group proteinsHMG1 and HMG2IBRDC2Zn-finger, RING; Zn-finger, cysteine-rich C6HC1.60.04ADIPOR1Receptor for globular and full-length adiponectin1.60.01(APM1), an essential hormone secreted by adipocytes.Probably involved in metabolic pathways that regulatelipid metabolism such as fatty acid oxidation.NDUFS7NADH-ubiquinone oxidoreductase 20 kDa subunit, mitochondrial1.60.01precursor; Complex I-20KD; CI-20KD; PSSTsubunitFGF6Can transform NIH 3T3 cells. Exhibits strong mitogenic1.60.02and angiogenic propertiesABHD5Alpha/beta hydrolase; Esterase/lipase/thioesterase, active1.60.04site; Prolyl aminopeptidase S33ABCC8Mono-heme cytochrome b. Regulator of ATP-sensitive1.60.06K+ channels and insulin releaseUFC1E2-like enzyme which forms an intermediate with UFM11.60.03via a thioester linkageQPCTResponsible for the biosynthesis of pyroglutamyl1.60.01peptides.KIAA0196Protein KIAA01961.60.04HMGA1HMG-I/Y bind preferentially to the minor groove of A + T1.60.05rich regions in double stranded DNA. Also involved intranscription regulation of genes containing, or near toA + T-rich regionsQ9BV99Leucine-rich repeat1.60.00RAPGEF2Guanine nucleotide exchange factor (GEF) for Rap1A1.60.02and Rap2B GTPases. It does not interact with cAMP orcGMPQ9BRP1Heat shock protein DnaJ, N-terminal; Programmed cell1.60.05death protein 2, C-terminalSPATA5L1AAA ATPase; ATP/GTP-binding site motif A (P-loop)1.60.06BDNFPromotes the survival of neuronal populations that are all1.60.04located either in the central nervous system or directlyconnected to it.C20orf85Protein C20orf851.60.04THBDThrombomodulin is a specific endothelial cell receptor1.60.04that forms a 1:1 stoichiometric complex with thrombin.This complex is responsible for the conversion of proteinC to the activated protein C (protein Ca).HSD3B13beta-HSD is a bifunctional enzyme, that catalyzes the1.60.04oxidative conversion of hormonal steroids andketosteroids.Q9BYH8Ankyrin1.60.00Q8WUE8Protein CGI-96 (PNAS-4).1.60.03ACRAcrosin is the major protease of mammalian1.60.03spermatozoa.Q8N7N1unknown1.60.01PRG1May neutralize hydrolytic enzymes1.60.02ZNF197ATP/GTP-binding site motif A (P-loop); Zn-finger,1.60.02C2H2 typeSERPINB1Regulates the activity of the neutrophil proteases1.60.03elastase, cathepsin G and proteinase-3EPHB2Receptor for members of the ephrin-B family1.60.01AKT2General protein kinase capable of phosphorylating1.60.05several known proteinsADMAM and PAMP are potent hypotensive and vasodilatator1.60.06agents.SUI1Necessary for scanning and involved in initiation site1.60.01selection. Probably involved in translationPPM1DRequired for the relief of p53-dependent checkpoint1.60.05mediated cell cycle arrest.ACTR1AComponent of a multi-subunit complex involved in1.60.00microtubule based vesicle motility. It is associated withthe centrosomeQ96MB3Protein kinase1.60.02IFITM1Implicated in the control of cell growth. Involved in the1.60.02transduction of antiproliferative and homotypic adhesionsignalsZDHHC4Probable palmitoyltransferase ZDHHC4;1.60.03Q9H8N7Zn-finger, C2H2 type1.60.06EPN3Epsin-3; EPS-15 interacting protein 31.60.05DNAJB6DnaJ homolog subfamily B member 6; Heat shock1.60.02protein J2Q96AG0Maternal tudor protein; Staphylococcus nuclease (SNase-1.60.01like)RFX1Regulatory factor essential for MHC class II genes1.60.06expression. Binds to the X boxes of MHC class II genes.GTF3C5Zn-finger, C2H2 type1.60.00RIN3Potential Ras effector protein. May function as a GEF by1.60.05exchanging bound GDP for free GTPRGS3Down-regulates G-protein-mediated release of inositol1.60.04phosphates and activation of MAP kinases.EBI3Cytokine receptor, common beta/gamma chain;1.60.02Fibronectin, type III; Long hematopoietin receptor,soluble alpha chainFGF20Neurotrophic factor that regulates central nervous1.60.01development and functionZNHIT1Bipartite nuclear localization signal; HIT Zn-finger1.50.05COL7A1Stratified squamous epithelial basement membrane1.50.05protein that form anchoring fibrils which may contributeto epithelial basement membrane organization andadherence by interacting with extracellular matrix (ECM)proteins e.g type IV collagenHERC2Cytochrome b5; Regulator of chromosome condensation,1.50.03RCC1.Q8IV48DNA-binding SAP; Exonuclease1.50.06SELT; SeltSelenoprotein T precursor1.50.01CCT2Molecular chaperone; assist the folding of proteins upon1.50.05ATP hydrolysis. Plays role, in vitro, in the folding ofactin and tubulinADAMTS20May play a role in tissue-remodeling process occurring1.50.03in both normal and pathological conditionsQ86X29Short-chain dehydrogenase/reductase SDR;1.50.03TNFR/CD27/30/40/95 cysteine-rich regionPRSS12Plays a role in neuronal plasticity and the proteolytic1.50.02action may → structural reorganizations associated withlearning & memoryCOPEThe coatomer is a cytosolic protein complex that binds to1.50.02dilysine motifs and reversibly associates with Golgi non-clathrin-coated vesicles, which further mediatebiosynthetic protein transport from the ER, via the Golgito the trans Golgi network.KIAA1036NM_0149091.50.02CPNE6May function in membrane trafficking. Exhibits calcium-1.50.05dependent phospholipid binding properties.ALS2CR3Amyotrophic lateral sclerosis 2 chromosomal region1.50.04candidate gene protein 3MPHOSPH10Component of the 60-80S U3 small nucleolar1.50.05ribonucleoprotein (U3 snoRNP). Required for the earlycleavages during pre-18S ribosomal RNA processingCAB39LCalcium-binding protein 39-like; Mo25-like protein1.50.02PAPOLBPolymerase that creates the poly(A) tail of mRNA.1.50.01THPOLineage-specific cytokine affecting the proliferation and1.50.04maturation of megakaryocytes from committedprogenitor cells. May be major physiological regulator ofcirculating plateletsC7orf16Inhibits protein phosphatase-2A and protein1.50.05phosphatase-1CAPZBF-actin capping proteins bind in a Ca(2+)-independent1.50.03manner to the fast growing ends of actin filaments(barbed end) thereby blocking the exchange of subunitsat these ends.ZNF124ATP/GTP-binding site motif A (P-loop); KRAB box;1.50.03Zn-finger, C2H2 typeRNU3IP2Component of a nucleolar small nuclear1.50.01ribonucleoprotein particle (snoRNP) thought toparticipate in the processing and modification of pre-ribosomal RNAANAPC5Component of the anaphase promoting1.50.04complex/cyclosome (APC/C), a cell cycle-regulatedubiquitin ligase that controls progression through mitosisand the G1 phase of the cell cycleENSG00000134490low complexity; signal peptide; transmembrane1.50.00PPP1R3CPutative phosphatase regulatory subunit1.50.01ZFP28KRAB box; Zn-finger, C2H2 subtype1.50.03ADCYAP1Stimulates adenylate cyclase in pituitary cells1.50.00FKBP8Has no PPIase/rotamase activity; Regulates myosin1.50.05phosphatase activity. Augments Ca2+sensitivity of thecontractile apparatusMTM1Dual-specificity phosphatase that acts on both1.50.06phosphotyrosine and phosphoserine. Could be involvedin a signal transduction pathway necessary for latemyogenesis, although its ubiquitous expression suggestsa wider functionQ7Z5U6G-protein beta WD-40 repeat1.50.02DKK1Inhibitor of Wnt signaling pathway1.50.02GALNT11Glycosyl transferase, family 2; Ricin B lectin domain1.50.03IL10Inhibits the synthesis of a number of cytokines, including1.50.03IFN-gamma, IL-2, IL-3, TNF and GM-CSF produced byactivated macrophages and by helper T cellsTGFBR1Receptor for TGF-beta. On ligand binding forms a1.50.01receptor complex consisting of two type II and two type Itransmembrane serine/threonine kinases, leads toactivation of SMAD TFs.DDX3XATP-dependent RNA helicase.1.50.01MRPL2Ribosomal protein L21.50.03BTRCSubstrate-recognition component of the SCF (SKP1-1.50.01CUL1-F-box protein) ubiquitin ligase complex, whichmediates the ubiquitination of proteins involved in cellcycle progression, signal transduction and transcription.Regulates the stability of CTNNB1 and participates inWnt signalingFGF10May be a growth factor active in the process of wound1.50.02healing. Acts as a mitogen in the lung, similar to FGF-7PDCD11Involved in the biogenesis of rRNA1.50.01O75250Cytochrome c heme-binding site1.50.05Wdr68WD-repeat protein 68; WD-repeat protein An111.50.03homologC12orf14HEJ1.1.50.04ATP5EThis is the smallest of the 5 chains of the enzymatic1.50.02component (coupling factor CF(1)) of the mitochondrialATPase complexHLA-DMAPlays a critical role in catalyzing the release of class II−1.50.02HLA-associated invariant chain-derived peptides (CLIP)from newly synthesized class II HLA molecules andfreeing the peptide binding site for acquisition ofantigenic peptidesENSG00000126970Cytochrome c heme-binding site−1.50.06CLECSF6C-type lectin; Type II antifreeze protein−1.50.04GIMAP4MSComponent of the 60-80S U3 small nucleolar−1.50.01ribonucleoprotein (U3 snoRNP). Required for the earlycleavages during pre-18S ribosomal RNA processing;Exhibits intrisinic GTPase activity.Q86XY4Tripin; shugoshin-like 2−1.50.02Q8IZY6Pleckstrin putative G-protein interacting domain;−1.50.04RhoGAP domainINHBEInhibins inhibit the secretion of follitropin by the−1.50.03pituitary gland.Q9HBJ8collectrin. kidney-specific membrane protein−1.50.01ALDH4A1Irreversible conversion of delta-1-pyrroline-5-−1.50.05carboxylate (P5C), derived either from proline orornithine, to glutamate. This is a necessary step in thepathway interconnecting the urea and tricarboxylic acidcycles.Q9NWR0Bipartite nuclear localization signal; Zn-finger, RING−1.50.04VDAC3Forms a channel through the mitochondrial outer−1.50.02membrane that allows diffusion of small hydrophilicmoleculesTHRBHigh affinity receptor for triiodothyronine−1.50.03PIK3CAPhosphorylates PtdIns, PtdIns4P and PtdIns(4,5)P2 with−1.50.00a preference for PtdIns(4,5)P2SOX11Probably important in the developing nervous system−1.50.01AKAP4Protein kinase A anchoring protein 4, 82 kDa−1.50.02Q9Y6Y3IDN3 protein isoform A.−1.50.06O75183coiled-coil; low complexity−1.50.04Q9H7R3Generic methyltransferase; SAM (and some other−1.50.02nucleotide) binding motifKIAA0084Hypothetical protein KIAA0084; HA2022−1.50.03Q9H7M9Immunoglobulin-like−1.50.04SLAMF8BCM-like membrane protein precursor. B lymphocyte−1.50.01activator macrophage expressedCORO1BMay be involved in cytokinesis, motility, and signal−1.50.03transductionCALM3Calmodulin mediates the control of a large number of−1.50.01enzymes by Ca(2+).MRPS6Mitochondrial 28S ribosomal protein S6; S6mt; MRP-S6−1.50.04FCGR3AReceptor for the Fc region of IgG. Binds complexed or−1.50.03aggregated IgG and also monomeric IgG. Mediatesantibody-dependent cellular cytotoxicity (ADCC) andother antibody-dependent responses, such asphagocytosisPOLQCould be involved in the repair of interstrand crosslinks−1.50.06ANKRD18AAnkyrin repeat domain protein 18A−1.50.01NUP54Component of the nuclear pore complex, a complex−1.50.03required for the trafficking across the nuclear membraneG6PCMay be a single membrane channel protein acting both−1.50.02as a hydrolase and a translocase. It is the key enzyme inhomeostatic regulation of blood glucose levelsZNF646May function as a transcription factor−1.50.05ZNF442KRAB box; Zn-finger, C2H2 subtype; Zn-finger, C2H2−1.50.04typeUBE2R2Ubiquitin-conjugating enzymes−1.50.00NPM3May act as a chaperone−1.50.02Q9H6X4NULL−1.50.03TFAMInvolved in mitochondrial transcription regulation as an−1.50.01activator Is able to unwind and bend DNAGDNFNeurotrophic factor that enhances survival and−1.50.04morphological differentiation of dopaminergic neuronsand increases their high-affinity dopamine uptakeMRPL24KOW; Ribosomal protein L24/L26−1.50.01C10orf45Bipartite nuclear localization signal−1.50.05FGD3DH domain; Pleckstrin-like; Zn-finger, FYVE type−1.50.01MBNL2Zn-finger, C-x8-C-x5-C-x3-H type−1.50.04NUCKS1Nuclear ubiquitous casein and cyclin-dependent kinases−1.50.06substrate; P1TBL1YF-box-like protein involved in the recruitment of the−1.50.01ubiquitin/19S proteasome complex to nuclear receptor-regulated transcription units.PCDHB14Potential calcium-dependent cell-adhesion protein. May−1.60.02be involved in the establishment and maintenance ofspecific neuronal connections in the brainPPM1FDephosphorylates and concomitantly deactivates CaM-−1.60.01kinases Promotes apoptosisENSG00000158142ATP/GTP-binding site motif A (P-loop); C2 domain−1.60.04Q9H7L1low complexity−1.60.00LUC7LProtein of unknown function DUF259−1.60.03UPF3BBipartite nuclear localization signal; Smg-4/UPF3−1.60.02DHCR24Catalyzes the reduction of the delta-24 double bond of−1.60.02sterol intermediates. Protects cells from oxidative stressby reducing caspase 3 activity during apoptosis inducedby oxidative stress.MKRN1Makorin-1; RING finger protein 61−1.60.01TUBCould be involved in the hypothalamic regulation of−1.60.05body weightENSG00000176783Cytochrome c heme-binding site; RUN domain; Zn-−1.60.03finger, C-x8-C-x5-C-x3-H type; Zn-finger, FYVE type;Zn-finger, RINGRTN3Reticulon-3; Neuroendocrine-specific protein-like 2;−1.60.03NSP-like protein II; NSPLIIQ8N8E1Apoptosis-related protein PNAS-1.−1.60.05Q8N283Ankyrin−1.60.03Q8IVV7low complexity−1.60.03Q7Z6C2A-kinase anchoring protein 95 (AKAP95); Bipartite−1.60.00nuclear localization signalQ8N570Parathyroid hormone-responsive osteosarcoma B1−1.60.01proteinALAS15-aminolevulinate synthase, nonspecific, mitochondrial−1.60.05precursor;MMP20Degrades amelogenin, the major protein component of−1.60.02the enamel matrix and two of the macromoleculescharacterising the cartilage extracellular matrixGALK2Acts on GalNAc. Also acts as a galactokinase when−1.60.01galactose is present at high concentrationsCENTG3GTPase-activating protein for the ADP ribosylation−1.60.03factor familyQ9HC06CD14 protein.−1.60.00PPM1AEnzyme with a broad specificity−1.60.03BMP15May be involved in follicular development.−1.60.01Q9P107Aldehyde dehydrogenase; Protein kinase C, phorbol−1.60.05ester/diacylglycerol binding; RhoGAP domainSPG4Probable ATPase involved in the assembly or function of−1.60.03nuclear protein complexes & maybe in aspects ofmicrotubule dynamicsNICALMay be a cytoskeletal regulator that connects NEDD9 to−1.60.03intermediate filamentsDEPDC5DEP domain containing protein 5−1.60.00Q7Z5B3RIC3 protein.−1.60.03MAGEH1Melanoma-associated antigen H1; Restin; Apoptosis-−1.60.06related protein 1; APR-1VDAC1Forms a channel through the mitochondrial outer−1.60.05membrane and also the plasma membrane; allowsdiffusion of small hydrophilic molecules.THRBHigh affinity receptor for triiodothyronine−1.60.01GDNFNeurotrophic factor that enhances survival and−1.60.03morphological differentiation of dopaminergic neuronsENSG00000188121Prenyl group binding site (CAAX box); Proline-rich−1.60.01extensin; Proline-rich regionPTGER2Receptor for prostaglandin E2 (PGE2).−1.60.05AREGBifunctional growth-modulating glycoprotein.−1.60.02HRMT1L3Probably methylates the guanidino nitrogens of arginyl−1.60.03residues in some proteinsRNF122Zn-finger, RING−1.60.00OSGEPGlycoprotease (M22) metalloprotease−1.60.01Q86VH4Leucine-rich repeat−1.60.04LOH12CR1LOH1CR12.−1.60.00STAT1Signal transducer and activator of transcription that−1.60.02mediates signaling by interferons (IFNs).BTBD7BTB/POZ domain−1.60.04TUSC2May function as a tumor suppressor, inhibiting colony−1.60.01formation, causing G1 arrest and ultimately inducingapoptosisQ8NF81low complexity−1.60.01TGFB1Tumour growth factor B1; Multifunctional peptide that−1.60.01controls proliferation, differentiation, and otherfunctions.SERPINB7Might function as an inhibitor of Lys-specific proteases.−1.60.02GRM1Receptor for glutamate.−1.60.02Q8TBK2Nuclear protein SET−1.60.02NT5C1B5′-nucleotidase, cytosolic IB; autoimmune infertility-−1.60.03related protein;UTRNMay play a role in anchoring the cytoskeleton to the−1.60.05plasma membrane (By similarity to dystrophin)PRKAG2AMPK is responsible for the regulation of fatty acid−1.60.03synthesis by phosphorylation of acetyl-CoA carboxylase.Q9H814coiled-coil; low complexity−1.60.01TCF7L2Participates in the Wnt signaling pathway and modulates−1.60.06MYC expression by binding to its promoter in asequence-specific manner.EFNB1Binds to the receptor tyrosine kinases EPHB1 and−1.60.00EPHA1.Q96LI9Bipartite nuclear localization signal−1.60.02Q8TBP6Mitochondrial carrier protein; Mitochondrial substrate−1.60.05carrierGPTParticipates in cellular nitrogen metabolism and in liver−1.60.02gluconeogenesisQ8N1Z9Like hepatocellular carcinoma-associated antigen−1.60.05HCA557b.ROBO2Fibronectin, type III; Immunoglobulin-like−1.60.02IL13RA1Binds IL13 with a low affinity. Together with IL4R-−1.60.04alpha can form a functional receptor for IL13. Alsoserves as an alternate accessory protein to the commonIL4 receptor gamma chainSVILATP/GTP-binding site motif A (P-loop); Bipartite−1.60.00nuclear localization signal; Gelsolin; Gelsolin region;Villin headpieceC6orf80Low complexity−1.60.02TMEM1May play role in vesicular transport from endoplasmic−1.60.03reticulum to GolgiMYO1BMotor protein that may participate in process critical to−1.60.03neuronal development and function such as cellmigration, neurite outgrowth and vesicular transportSV2BGeneral substrate transporter; Sugar transporter−1.60.02superfamilyVCLInvolved in cell adhesion. May be involved in the−1.60.01attachment of the actin-based microfilaments to theplasma membraneCBX5Component of heterochromatin. Recognizes and binds−1.60.03histone H3 tails methylated at Lys-9, leading toepigenetic repression.AKR1B10Can efficiently reduce aliphatic and aromatic aldehydes,−1.60.03and is less active on hexoses.MRPL1Ribosomal protein L1−1.60.04Q9BSA9NULL−1.60.05ABCD3Probable transporter. The nucleotide-binding fold acts as−1.60.04an ATP-binding subunit with ATPase activityFMODAffects the rate of fibrils formation. May have a primary−1.60.04role in collagen fibrillogenesisGRIP1PDZ/DHR/GLGF domain−1.60.03TMEM22Protein of unknown function DUF6−1.60.02ZFYVE21Zn-finger, FYVE type−1.60.03SPPL2AMay act as intramembrane protease−1.60.04CUGBP2Paraneoplastic encephalomyelitis antigen; RNA-binding−1.60.01region RNP-1Q96IW2SH2 motif−1.60.02C20orf147Haloacid dehalogenase-like hydrolase domain containing−1.60.05prot. 4Q8N2K3low complexity; transmembrane−1.70.06Q9BSD4coiled-coil; low complexity−1.70.03LAMC1Binding to cells via a high affinity receptor, laminin is−1.70.02thought to mediate the attachment, migration andorganization of cells into tissues during embryonicdevelopment by interacting with other extracellularmatrix componentsQ8NEH9IQ calmodulin-binding region−1.70.04SEMA5BMay act as positive axonal guidance cues−1.70.00TMEM14ATransmembrane protein 14A−1.70.06ANGPTL1Fibrinogen, beta/gamma chain, C-terminal globular−1.70.00Q9NSN6TPR repeat−1.70.01PARNDeadenylation nuclease; poly(A)-specific ribonuclease−1.70.03NPY6RNeuropeptide Y receptor; Rhodopsin-like GPCR−1.70.01superfamilyQ9H089ATP/GTP-binding site motif A (P-loop)−1.70.05SKP1AEssential component of the SCF (SKP1-CUL1-F-box−1.70.03protein) ubiquitin ligase complexCNOT7Ubiquitous transcription factor required for a diverse set−1.70.02of processes. Component of the CCR4 complex.MYO1CMyosins are actin-based motor molecules with ATPase−1.70.04activity. Unconventional myosins serve in intracellularmovement.PRKCBP1Protein kinase C binding protein 1; Rack7; Cutaneous T-−1.70.03cell lymphoma associated antigen sel4-3;SH3BGRL3Could act as a modulator of glutaredoxin biological−1.70.01activityMTHFRCatalyzes the conversion of 5,10-−1.70.01methylenetetrahydrofolate to 5-methyltetrahydrofolate,for remethylation to methionineNUP155Essential component of nuclear pore complex.−1.70.04SMARCA3Helicase, C-terminal; SNF2 related domain; Zn-finger,−1.70.02RINGSH3MD3SH3 domain−1.70.03GSTM1Conjugation of reduced glutathione to a wide number of−1.70.03exogenous and endogenous hydrophobic electrophilesC20orf140May act as a GTPase activating protein for Rab family−1.70.03protein(s)TAGLN3Transgelin-3; Neuronal protein NP25; Neuronal protein−1.70.0422; NP22Q96B77transmembrane−1.70.04OR52A1Putative odorant receptor−1.70.03ACTN1F-actin cross-linking protein which is thought to anchor−1.70.00actin to a variety of intracellular structures. This is abundling proteinSLC38A4Amino acid/polyamine transporter, family II−1.70.05APPFunctions as a cell surface receptor and can promote−1.70.03transcription activation through binding to APBB1/Tip60and inhibit Notch signaling through interaction withNumb. Couples to apoptosis-inducing pathwaysQ96CE7FAD-dependent pyridine nucleotide-disulphide−1.70.01oxidoreductase; Flavin-containing monooxygenase(FMO) 1;COQ4Ubiquinone biosynthesis protein COQ4 homolog;−1.70.05Coenzyme Q biosynthesis protein 4 homologFAF1Potentiates but cannot initiate FAS-induced apoptosis−1.70.03Q9NTC3Eukaryotic/viral aspartic protease, active site−1.70.06ARPC1APart of a complex implicated in the control of actin−1.70.02polymerization in cellsTRPM4Ion transport protein−1.70.04P2RY5P2Y purinoceptor 5; P2Y5; Purinergic receptor 5; RB−1.70.01intron encoded G-protein coupled receptorTLK2Rapidly and transiently inhibited by phosphorylation−1.80.02following the generation of DNA double-stranded breaksduring S-phase.COMMD3BUP protein; chromosome 10 open reading frame 8.−1.80.01COMM domain containing 3ELA3AEfficient protease with alanine specificity but only little−1.80.04elastolytic activityKIAA0574Hypothetical protein KIAA0574−1.80.03UBE1LActivates ubiquitin.−1.80.03Q8TB55Proline-rich region−1.80.05RAB28Ras-related protein Rab-28; Rab-26−1.80.03ASB8Ankyrin repeat and SOCS box protein 8; ASB-8−1.80.01SNX3May be involved in several stages of intracellular−1.80.02traffickingJUNDBinds an AP-1 site and upon cotransfection stimulates−1.80.05the activity of a promoter that bears an AP-1 siteQ8NF73G-protein beta WD-40 repeat−1.80.03NUP43May mediate the assembly of subdomains of the NPC or−1.80.04facilitate the interaction of transport complexes with theNPCDCXSeems to be required for initial steps of neuronal−1.80.00dispersion and cortex lamination during cerebral cortexdevelopment.ASPHAspartyl/Asparaginyl beta-hydroxylase, N-terminal−1.80.03RPS6KA5Serine/threonine kinase that may play a role in mediating−1.80.05the growth-factor and stress induced activation of thetranscription factor CREB. Essential role in the controlof RELA transcriptional activity in response to TNFDNTTIP1Shown to enhance TdT activity, in vitro−1.80.00ZNF436May be involved in transcriptional regulation−1.80.02Q9NX40ovarian carcinoma immunoreactive antigen.−1.80.04HAGHThiolesterase that catalyzes the hydrolysis of S-D-−1.80.01lactoyl-glutathione to form glutathione and D-lactic acidMGEA6Tumor-associated antigen−1.80.01SLC19A1Transporter for the intake of folate.−1.80.01SULT4A1May catalyze the sulfate conjugation of many drugs,−1.80.05xenobiotic compounds, hormones, and neurotransmitters.NAV1ATP/GTP-binding site motif A (P-loop); Bipartite−1.80.03nuclear localization signal; Inorganic pyrophosphataseANGPTL6Fibrinogen, beta/gamma chain, C-terminal globular−1.80.03NDUFA5Transfer of electrons from NADH to the respiratory−1.80.04chain. This is a component of the iron-sulfur (IP)fragment of the enzymeC6orf37low complexity−1.80.05C9orf86ATP/GTP-binding site motif A; Ras GTPase superfamily−1.80.04Q8NAA4G-protein beta WD-40 repeat−1.80.00CETN2Plays a fundamental role in microtubule-organizing−1.90.01center structure and functionCEECAM1Endoplasmic reticulum targeting sequence; Glycosyl−1.90.03transferase, family 25USP6NLRabGAP/TBC domain−1.90.01FOLH1Has both folate hydrolase and N-acetylated-alpha-linked-−1.90.01acidic dipeptidase (NAALADase) activity. Involved inprostate tumor progressionCYP51A1Catalyzes C14-demethylation of lanosterol.−1.90.05ITGB6Integrin alpha-V/beta-6 is a receptor for fibronectin and−1.90.06cytotactin. It recognizes the sequence R-G-D in itsligandsRIPK3Promotes apoptosis−1.90.05Q8TDG4DEAD/DEAH box helicase; Helicase, C-terminal−1.90.03ALS2CR3Amyotrophic lateral sclerosis 2 chromosomal region−1.90.02candidate gene protein 3VMD2L3Forms calcium-sensitive chloride channels. May conduct−1.90.04other physiologically significant anions such asbicarbonateO95893transmembrane−1.90.03SNTB1Adapter protein that binds to and probably organizes the−1.90.04subcellular localization of a variety of membraneproteins. May link various receptors to the actincytoskeletonMYPNEndoplasmic reticulum targeting sequence;−1.90.02Immunoglobulin-likeQ8WTU5ATP/GTP-binding site motif A (P-loop)−1.90.01ECHDC1Enoyl-CoA hydratase/isomerase−1.90.02Q96JT2Acc: NM_033102]; prostein protein. [Source: RefSeq−1.90.04Q9BZS9Acc: Q9BZS9]; PNAS-138. [Source: SPTREMBL−1.90.00Q86UX6Protein kinase; Serine/Threonine protein kinase−1.90.02SCN9AATP/GTP-binding site motif A (P-loop); Cation channel,−1.90.04non-ligand gated; IQ calmodulin-binding region;Polycystic kidney disease type 2 proteinBLVRBCatalyzes electron transfer from reduced pyridine−1.90.03nucleotides to flavins. Possible role in protecting cellsfrom oxidative damage or in regulating iron metabolism.KIFAP3Involved in tethering the chromosomes to the spindle−2.00.01pole and in chromosome movement.USP9YMay function as a ubiquitin-protein or polyubiquitin−2.00.01hydrolase.PAK4Activates the JNK pathway.−2.00.02SLC39A1Mediates zinc uptake. May function as a major−2.00.01endogenous zinc uptake transporter in many cells of thebody.Q8NCL8low complexity; signal peptide; transmembrane−2.00.03Q8NA48testes development-related NYD-SP18.−2.00.00Q96CY3Bipartite nuclear localization signal−2.00.04JUBSugar transporter superfamily; Zn-binding protein, LIM−2.00.02CSF1RReceptor for CSF-1, protein tyrosine-kinase−2.00.01ATP7BInvolved in the export of copper out of the cells, such as−2.00.02the efflux of hepatic copper into the bileKIAA1244Essential component of the high affinity receptor for the−2.00.03general membrane fusion machinery and an importantregulator of transport vesicle docking and fusionHTR1DOne of the several different receptors serotonin.−2.00.01C9orf114Bipartite nuclear localization signal; DUF171−2.00.01HIF3ABasic helix-loop-helix dimerization domain bHLH;−2.00.01Nuclear translocator; PAS domainARL14Involved in protein trafficking; may modulate vesicle−2.00.01budding and uncoating within the Golgi apparatusMEF2CTranscription activator which binds specifically to the−2.10.01MEF2 element in the regulatory regions of many muscle-specific genes.Q9P1V9low complexity−2.10.05FRMPD1PDZ/DHR/GLGF domain; RA domain−2.10.05O60592Neutrophil cytosol factor 2; Proline-rich extensin; SH3−2.10.02domain; Sorbin-like; Zn-finger, C2H2 typeC6orf65coiled-coil−2.10.06ASB4Ankyrin repeat and SOCS box protein 4; ASB-4−2.10.02Q8IY68low complexity−2.10.06Q9BU59G-protein beta WD-40 repeat−2.10.04COX5BOne of the nuclear-coded polypeptide chains of−2.20.01cytochrome c oxidase, the terminal oxidase inmitochondrial electron transportCOL4A3BPPhosphorylates on Ser and Thr residues the Goodpasture−2.20.03autoantigen (in vitro). Isoform 2 seems to be less activeRRHMay play a role in rpe physiology either by detecting−2.20.01light directly or by monitoring the concentration ofretinoids or other photoreceptor-derived compoundsQ8TCQ1Bipartite nuclear localization signal; Zn-finger, RING−2.30.04KLRG1C-type lectin−2.30.05ZNF385Zn-finger, C2H2 matrin type; Zn-finger, C2H2 type−2.30.05VAT1Synaptic vesicle membrane protein VAT-1 homolog−2.30.03C14orf161transmembrane−2.30.06HK1Hexokinase type I; HK I; Brain form hexokinase−2.40.02EEDG-protein beta WD-40 repeat; Regulator of chromosome−2.40.04condensation, RCC1C22orf3Protein C22orf3−2.60.04PPT1Removes thioester-linked fatty acyl groups such as−3.00.04palmitate from modified cysteine residues in proteins orpeptides during lysosomal degradation.SCGB2A2Mammaglobin A precursor; Mammaglobin-1;−3.10.02Secretoglobin family 2A member 2O75915Prenylated rab acceptor PRA1−3.40.00Q86VG1Bipartite nuclear localization signal; NF-X1 type; Zn-−4.70.04finger, RING









TABLE 71










Gene profiling of differentially expressed genes in human monocytes due to the


presence of bacterial endotoxin (LPS) and SEQ ID NO: 7 revealing 1012 differentially


expressed genes.












Fold





Change by


Gene Name
Gene Description
LPS + SEQ 7
p-value













RBP1
Intracellular transport of retinol
125.8
0.05


TMOD4
Blocks the elongation and depolymerization of the actin
115.9
0.04



filaments at the pointed end.


Q8WUC6
Bipartite nuclear localization signal; Class I peptide
104.7
0.05



chain release factor domain


GPD1
Glycerol-3-phosphate dehydrogenase [NAD+],
89.7
0.04



cytoplasmic


KCNH7
Pore-forming α-subunit of voltage-gated potassium
80.6
0.06



channel. Channel properties may be modulated by cAMP


O43300
Leucine-rich repeat
77.1
0.03


TGM4
Associated with the mammalian reproductive process.
73.5
0.04



Catalyzes the cross-linking of proteins and the



conjugation of polyamines to specific proteins in the



seminal tract


POU1F1
Transcription factor involved in the specification of the
64.2
0.03



lactotrope, somatotrope, and thyrotrope phenotypes in



the developing anterior pituitary. Activates growth



hormone and prolactin genes. Specifically binds to the



consensus sequence 5′-TAAAT-3′;


FOXP1
Transcriptional repressor that play an important role in
60.4
0.04



the specification and differentiation of lung epithelium


KCNK6
Exhibits outward rectification in a physiological K(+)
58.8
0.05



gradient and mild inward rectification in symmetrical



K(+) conditions


Q9C098
Protein kinase; Serine/Threonine protein kinase
57.1
0.01


RHBDF1
Rhomboid-like protein
54.7
0.04


Q8N135
ATP/GTP-binding site motif A (P-loop);
53.9
0.06



PDZ/DHR/GLGF domain


CD226
Immunoglobulin-like
53.2
0.03


O43348
Argininosuccinate synthase
51.7
0.02


SMF
SMF protein
51.1
0.05


Q9Y4T9
low complexity
48.3
0.05


Q86WW9
ATP/GTP-binding site motif A (P-loop); Lipoxygenase,
47.2
0.02



LH2 domain


DLX5
Homeobox protein DLX-5
43.2
0.05


SMURF2
E3 ubiquitin-protein ligase which accepts ubiquitin from
42.1
0.04



an E2 ubiquitin-conjugating enzyme in the form of a



thioester and then directly transfers the ubiquitin to



targeted substrates. Interacts with SMAD1, SMAD2 and



SMAD7 in order to trigger their ubiquitination and



proteasome-dependent degradation.


CNTN5
Fibronectin, type III; Immunoglobulin-like
40.5
0.05


ZNF73
ATP/GTP-binding site motif A (P-loop); KRAB box;
36.4
0.03



Zn-finger, C2H2 subtype;


ARNT
Required for activity of the Ah (dioxin) receptor. This
35.7
0.05



protein is required for the ligand-binding subunit to



translocate from the cytosol to the nucleus after ligand



binding.


BNIP1
Implicated in the suppression of cell death. Interacts with
34.9
0.04



the BCL-2 and adenovirus E1B 19 kDa proteins


ITGA8
Integrin alpha-8/beta-1 is a receptor for fibronectin and
34.0
0.06



cytotactin. It recognizes the sequence R-G-D in its



ligands


ZNF302
May function as a transcription factor
33.6
0.04


GFER
Augmenter of liver regeneration (hERV1 protein).
32.7
0.03


NR2F2
Regulation of the apolipoprotein A-I gene transcription.
32.7
0.03



Binds to DNA site A


STATH
Salivary protein that stabilizes saliva supersaturated with
31.8
0.03



Ca2+ salts by inhibiting the precipitation of calcium



phosphate salts.


Q9H697
limkain beta 2.
29.4
0.05


IDUA
Alpha-L-iduronidase precursor
28.7
0.03


WBSCR18
Williams-Beuren syndrome chromosome region 18
27.4
0.06



protein


DNAJC1
DnaJ homolog subfamily C member 1
26.9
0.05


NRXN1
Neuronal cell surface protein that may be involved in
25.9
0.04



cell recognition and cell adhesion. May mediate



intracellular signaling


FEZ2
Involved in axonal outgrowth and fasciculation
25.8
0.02


OR5U1
Putative odorant receptor
23.8
0.04


ENSG00000162701
DENN (AEX-3) domain; uDENN domain
22.8
0.04


MCART1
Mitochondrial carrier triple repeat 1
22.8
0.05


SBNO1
Helicase, C-terminal; RNA-binding region RNP-1
22.2
0.05


CHRNA1
Acetyl choline receptor. After binding acetylcholine, the
21.3
0.03



AChR leads to opening of an ion-conducting channel



across the plasma membrane


HOOK2
Probable cytoskeletal linker protein, which may be
19.9
0.02



involved in tethering membrane bound organelles to the



cytoskeleton


ENSG00000105849
RNA polymerase Rpa43 subunit
19.8
0.05


Q8N6Q6
unknown
18.8
0.04


FHL3
Four and a half LIM domains protein 3; FHL-3; Skeletal
18.4
0.02



muscle LIM-protein 2; SLIM 2


CKMT1
Reversibly catalyzes the transfer of phosphate between
17.8
0.04



ATP and various phosphogens (e.g. creatine phosphate).



Creatine kinase isoenzymes play a central role in energy



transduction.


Q8N8U9;
Trypsin inhibitor-like, cysteine-rich TIL region; Vitamin
17.7
0.05


Q8TF36
K-dependent carboxylation/gamma-carboxyglutamic



(GLA) domain; von Willebrand factor, type C, D


BLZF1
basic leucine zipper nuclear factor 1.
17.6
0.05


Q9BRK2
Protein of unknown function DUF625
17.6
0.04


IDH3G
Isocitrate dehydrogenase [NAD] subunit gamma,
17.5
0.04



mitochondrial precursor;


IL17C
Stimulates the release of tumor necrosis factor alpha and
17.4
0.05



IL-1 beta from the monocytic cell line THP-1


Q9H6R7
Coiled-coil; low complexity
17.1
0.06


OR5P2
Putative odorant receptor. Could also be involved in taste
17.1
0.05



perception


PLCG1
phospholipase C-gamma is a major substrate for heparin-
17.0
0.04



binding growth factor 1 (acidic fibroblast growth factor)-



activated tyrosine kinase


Q8NHU6
Bipartite nuclear localization signal; Maternal tudor
16.4
0.04



protein


RNF24
RING finger protein 24
16.2
0.03


Q9H9X6
Bipartite nuclear localization signal
16.0
0.05


MAP3K1
Component of a protein kinase signal transduction
15.9
0.04



cascade. Activates the ERK and JNK kinase pathways by



phosphor-ylation of MAP2K1 and MAP2K4. Activates



CHUK and IKBKB, the central protein kinases of the



NFκB pathway


ALDOB
Fructose-bisphosphate aldolase B; Liver-type aldolase
15.5
0.01


Q96LW2
Blue (type 1) copper domain; Protein kinase;
15.3
0.04



Serine/Threonine protein kinase


EPB41L4B
Band 4.1-like protein 4B; EHM2 protein; FERM-
13.7
0.04



containing protein CG1


RCL1
Plays a role in 40S-ribosomal-subunit biogenesis in the
13.5
0.01



early pre-rRNA processing steps at sites A0, A1 and A2



that are required for proper maturation of the 18S RNA


PTGDS2;
Catalyzes the conversion of PGH2 to PGD2, a
13.3
0.04


PGDS
prostaglandin that is a potent inhibitor of platelet



aggregation


RGS14
Inhibits signal transduction by increasing GTPase
13.1
0.05



activity of G protein α-subunits driving them into



inactive GDP-bound form


CYLC2
Cylicin II (Multiple-band polypeptide II).
12.7
0.03


GPR174
Putative receptor for purines coupled to G-proteins
12.4
0.04


PMPCB
Cleaves presequences (transit peptides) from
12.3
0.05



mitochondrial protein precursors


ANKRD5
Ankyrin repeat domain protein 5
12.2
0.05


SYNE1
Involved in the maintenance of nuclear organization and
12.2
0.05



structural integrity. Probable anchoring protein which



theters the nucleus to the cytoskeleton. Connects nuclei



to the cytoskeleton by interacting with the nuclear



envelope and with F-actin in the cytoplasm


IL17B
Stimulates the release of tumor necrosis factor alpha and
12.2
0.02



IL-1 beta from the monocytic cell line THP-1


GTF2H1
Component of the core-TFIIH basal transcription factor
12.1
0.04



involved in nucleotide excision repair (NER) of DNA



and, in complex with CAK, in transcription by RNA



polymerase II


ARRB1
Regulates beta-adrenergic receptor function.
12.0
0.03


RAB22A
Ras-related protein Rab-22A; Rab-22; Rab-31; Rab-22B
11.9
0.04


OSBPL7
Oxysterol binding protein-related protein 7; ORP-7
11.9
0.01


FBN1
Fibrillins are structural components of 10-12 nm
11.8
0.03



extracellular calcium-binding microfibrils, which occur



either in association with elastin or in elastin-free



bundles.


PTGER3
Receptor for prostaglandin E2 (PGE2)
11.8
0.03


SIRT1
NAD-dependent deacetylase, which regulates processes
11.8
0.04



such as apoptosis and muscle differentiation by



deacetylating key proteins.


STUB1
TPR repeat; Zn-finger, modified RING
11.8
0.05


DUSP14
Involved in the inactivation of MAP kinases.
11.6
0.01



Dephosphorylates ERK, JNK and p38 MAP-kinases


KRTHA4
Keratin, type I cuticular Ha4; Hair keratin, type I Ha4
11.4
0.00


SACM1L
Synaptojanin, N-terminal
10.9
0.06


Q8N336
Protein of unknown function DUF609
10.1
0.04


O60384
Zn-finger, C2H2 type
9.9
0.05


PTPRCAP
Protein tyrosine phosphatase receptor type C-associated
9.7
0.04



protein; Lymphocyte phosphatase-associated



phosphoprotein


UBE2N
The UBE2V2/UBE2N heterodimer catalyzes the
9.6
0.04



synthesis of non-canonical poly-ubiquitin chains that are



linked through Lys-63; doesn't lead to protein



degradation by the proteasome. Mediates transcriptional



activation of target genes. Plays a role in the control of



progress through the cell cycle and differentiation.


Q8TDS9
putative G-protein coupled receptor GPCR42.
9.6
0.00


LNX
E3 Ubiquitin ligase protein that mediates ubiquitination
9.5
0.06



and subsequent proteasomal degradation of NUMB.


GSTZ1
Bifunctional enzyme showing minimal glutathione-
9.3
0.05



conjugating activity and low glutathione peroxidase



activity


SLC27A6
AMP-dependent synthetase and ligase
9.2
0.05


CNTN6
ABC transporter; Fibronectin, type III; Immunoglobulin-
9.2
0.00



like


MSX1
Acts as a transcriptional repressor. May play a role in
9.2
0.05



limb-pattern formation. Acts in cranofacial development



and specifically in odontogenesis


Q8N4J6
HMG-I and HMG-Y DNA-binding domain (A + T-hook);
9.1
0.05



Pistil-specific extensin-like protein; Proline-rich extensin


MYL4
Regulatory light chain of myosin. Does not bind calcium
8.9
0.05


ARF1
GTP-binding protein involved in protein trafficking
8.8
0.01



among different compartments. Modulates vesicle



budding and uncoating within the Golgi complex.


Q8NHE2
SF21 protein.
8.7
0.04


Q8N3K5
Cysteine-rich flanking region, N-terminal;
8.7
0.06



Immunoglobulin-like; Leucine-rich repeat; RNA-binding



region RNP-1


Q9H5P1
Zn-finger, C-x8-C-x5-C-x3-H type
8.6
0.02


CDK7
Cyclin-dependent kinase-7; CDK7 is the catalytic
8.6
0.05



subunit of the CDK-activating kinase complex, a serine-



threonine kinase. Involved in cell cycle control and in



RNA transcription by RNA polymerase II.


Q7RTU0
Basic helix-loop-helix dimerization domain bHLH
8.4
0.06


ZNF322B
Zn-finger, C2H2 type
8.3
0.05


MPP4
May play a role in retinal photoreceptors development
8.2
0.03


ALOX5
Arachidonate 5-lipoxygenase; 5-lipoxygenase; 5-LO
8.1
0.03


NSF
May participate in trafficking events that are associated
8.0
0.06



with myogenesis, such as myoblast fusion and/or



GLUT4 trafficking; Required for vesicle-mediated



transport.


Q9NZ13
Zn-finger, C2H2 type
8.0
0.03


PPAP2B
PA-phosphatase related phosphoesterase
7.9
0.05


TUBGCP6
Gamma-tubulin complex is necessary for microtublule
7.8
0.03



nucleation at the centrosome


Q9BUJ0
Alpha/beta hydrolase; Esterase/lipase/thioesterase, active
7.6
0.04



site


TRPM3
Calcium channel mediating constitutive calcium ion
7.5
0.02



entry. Its activity is increased by reduction in



extracellular osmolarity, by store depletion and



muscarinic receptor activation


Q96E44
Beta and gamma crystallin; Nuclear protein SET
7.4
0.06


Q9P1G1
signal peptide
7.4
0.06


CENTB2
GTPase-activating protein for ADP ribosylation factor
7.4
0.05



family


XRCC5
Single stranded DNA-dependent ATP-dependent
7.3
0.03



helicase. Has a role in chromosome translocation.


GIT2
GTPase-activating protein for the ADP ribosylation
7.2
0.05



factor family


Q9BYE9
Cadherin
6.9
0.05


C15orf15
Bipartite nuclear localization signal; Ribosomal protein
6.9
0.04



L24E


RAD52
Involved in double-stranded break repair. Plays a central
6.9
0.05



role in genetic recombination and DNA repair


UBXD2
UBX domain-containing protein 2
6.8
0.04


Q86TW0
Bipartite nuclear localization signal; Zn-finger, C-x8-C-
6.8
0.04



x5-C-x3-H type


TCN1
Vitamin B12-binding protein. Transports cobalamin into
6.7
0.03



cells


CCS
Delivers copper to copper zinc superoxide dismutase
6.7
0.05



(SOD1)


Q96CN5
Leucine-rich repeat
6.6
0.04


FBXW5
Cyclin-like F-box; G-protein beta WD-40 repeat
6.5
0.05


C21orf108
Nucleolar preribosomal-associated protein 1
6.3
0.04


SOX9
Plays an important role in the normal skeletal
6.1
0.05



development. May regulate the expression of other genes



by acting as a transcription factor for these genes


SYT1
May have a regulatory role in the membrane interactions
6.1
0.06



during trafficking of synaptic vesicles at the active zone



of the synapse; binds acidic phospholipidsand can bind



to at least three additional proteins, neurexins, syntaxin



and AP2


SYT11
May be involved in Ca(2+)-dependent exocytosis of
6.1
0.06



secretory vesicles through Ca(2+) and phospholipid



binding to the C2 domain or may serve as Ca(2+)



sensors in the process of vesicular trafficking and



exocytosis


MPHOSPH6
M-phase phosphoprotein 6
6.0
0.03


ZNF208
KRAB box; Neutral zinc metallopeptidases, zinc-binding
6.0
0.04



region; Zn-finger, C2H2 subtype


HNRPH2
This protein is a component of the heterogenous nuclear
6.0
0.05



ribonucleoprotein (hnRNP) complexes


C13orf1
SPla/RYanodine receptor SPRY
5.9
0.04


NRG1
Direct ligand for ERBB3 and ERBB4 tyrosine kinase
5.8
0.02



receptors. The multiple isoforms perform diverse



functions such as inducing growth and differentiation of



epithelial, glial, neuronal, and skeletal muscle cells;


EIF2C4
Plays an important role in the eukaryotic peptide chain
5.7
0.05



initiation process


HOOK1
Cytoskeletal linker protein, which may be involved in
5.7
0.02



tethering membrane-bound organelles to the



cytoskeleton.


REPS1
May coordinate the cellular actions of activated EGF
5.6
0.04



receptors and Ral-GTPases


HLA-J
Immunoglobulin-like; Immunoglobulin/major
5.6
0.03



histocompatibility complex (MHC); MHC protein, class I


SYNGR2
Synaptogyrin-2; Cellugyrin
5.6
0.05


GRTP1
RabGAP/TBC domain; Somatotropin hormone
5.6
0.05


RNF41
Zn-finger, RING
5.5
0.03


PTGIS
Catalyzes the isomerization of prostaglandin H2 to
5.5
0.01



prostacyclin (=prostaglandin I2)


TFEC
Basic helix-loop-helix dimerization domain bHLH
5.5
0.01


C20orf108
Protein C20orf108
5.5
0.03


RPS7
40S ribosomal protein 57; 405 ribosomal protein S7; 58
5.4
0.04


ANKMY1
Ankyrin repeat and MYND domain protein 1; Testis-
5.4
0.00



specific ankyrin-like protein 1; Zinc-finger MYND



domain protein 13


GUCY1B3
Guanylate cyclase soluble, beta-1 chain; GCS-beta-1;
5.4
0.01



Soluble guanylate cyclase small subunit; GCS-beta-3


TIRAP
Adapter involved in the TLR4 signaling pathway in the
5.4
0.01



innate immune response. Acts via IRAK2 and TRAF-6,



leading to the activation of NF-kappa-B, MAPK1,



MAPK3 and JNK, resulting in cytokine secretion and the



inflammatory response


MRPS22
Mitochondrial 28S ribosomal protein S22; S22mt; MRP-
5.3
0.04



S22


IFNA2
Produced by macrophages, IFN-alpha have antiviral
5.3
0.01



activities. Interferon stimulates the production of two



enzymes: a protein kinase and an oligoadenylate



synthetase


MUC11
Actin-binding, actinin-type; Eukaryotic RNA
5.3
0.02



polymerase II heptapeptide repeat


KCNA6
Mediates the voltage-dependent potassium ion
5.2
0.06



permeability of excitable membranes.


SPG6
WW/Rsp5/WWP domain
5.2
0.03


Q96MA7
coiled-coil; low complexity
5.1
0.03


CCL20
Chemotactic factor that attracts lymphocytes and,
5.1
0.00



slightly, neutrophils, but not monocytes.


C20orf26
Protein C20orf26
5.1
0.01


TG
Precursor of the iodinated thyroid hormones thyroxine
5.0
0.05



(T4) and triiodothyronine (T3)


PAK2
The activated kinase phosphorylates a variety of targets,
5.0
0.06



e.g. ribosomal protein S6, histone H4 and myelin basic



protein.


TREX2
26S proteasome-associated UCH37 interacting protein 1;
4.9
0.02



X-linked protein STS1769


TMSB10
Plays an important role in the organization of the
4.9
0.02



cytoskeleton. Binds to and sequesters actin monomers (G



actin) and therefore inhibits actin polymerization


Q9BRX9
G-protein beta WD-40 repeat
4.9
0.04


Q9NW81
Leucine-rich repeat
4.8
0.02


FOXQ1
Forkhead box protein Q1; Hepatocyte nuclear factor 3
4.8
0.01



forkhead homolog 1;


PCCB
Propionyl-CoA carboxylase beta chain, mitochondrial
4.7
0.03



precursor;


Q9H7Y2
low complexity
4.7
0.03


PMAIP1
Phorbol-12-myristate-13-acetate-induced protein 1;
4.7
0.00



Immediate-early-response protein APR


SNRPC
This protein is associated with snRNP U1
4.6
0.02


Q969S1
Mitochondrial substrate carrier
4.5
0.01


BAZ2A
May play a role in transcriptional regulation interacting
4.5
0.01



with ISWI. May serve a specific role in maintaining or



altering the chromatin structure of the rDNA locus


DACH2
Bipartite nuclear localization signal; Transforming
4.5
0.04



protein Ski


Wdr68; Han11
WD-repeat protein 68; WD-repeat protein An11
4.5
0.05



homolog


CCL23
Shows chemotactic activity for monocytes, resting T-
4.5
0.05



lymphocytes, and neutrophils, but not for activated



lymphocytes.


ARHGEF1
Seems to play a role in the regulation of RhoA GTPase
4.4
0.00



by guanine nucleotide-binding alpha-12 (GNA12) and



alpha-13 (GNA13) subunits. Acts as GTPase-activating



protein (GAP) for GNA12 and GNA13, and as guanine



nucleotide exchange factor (GEF) for RhoA GTPase.


Q7Z620
C2 domain
4.4
0.04


MGAT5B
Beta(1,6)-N-acetylglucosaminyltransferase V isoform 1
4.3
0.05


BATF
Functions as negative regulator of AP-1 mediated
4.3
0.02



transcription by binding to Jun proteins. Jun/B-ATF



heterodimers bind DNA preferentially at the 12-O-



tetradecanoylphorbol-13-acetate response element (TRE)



(consensus: 5‘TGA[CG]TCA-3’) and weaker at the



cAMP responsive region (CRE) (consensus:



5‘GTGACGT[AC][AG]-3’), but are transcriptionally



inert


DF; PALM
May be involved in control of cell shape
4.3
0.02


SP3
Binds to GT and GC boxes promoters elements.
4.2
0.02



Probable transcriptional activator


DNMT2
Its strong binding to DNA suggests that it may mark
4.2
0.05



specific sequences in the genome by binding to DNA



through the specific target-recognizing motif. Doesn't



seem to be active as a DNA methyltransferase.


Q9NX89
unknown
4.1
0.04


Q96AF2
Protein kinase; Tyrosine protein kinase
4.1
0.00


CHCHD5
Bipartite nuclear localization signal
4.1
0.02


Q9NXD2
Bipartite nuclear localization signal
4.1
0.01


STIM1
Possible adhesion molecule with a role in early
4.1
0.03



hematopoiesis by mediating attachment to stromal cells.



Influences the survival and/or proliferation of B cell



precursors. Binding to cells requires Mn(2+)


OCLN; RPS27
May play a role in the formation and regulation of the
4.1
0.03



tight junction (TJ) paracellular permeability barrier


TNFSF5IP1
Tumor necrosis factor superfamily, member 5-induced
4.0
0.00



protein 1; HDCMC29P; HSPC260.;


Q96MX1
down-regulated by Ctnnb1, a.
4.0
0.03


TDRD1
Tudor domain-containing protein 1
4.0
0.04


C13orf11
coiled-coil; low complexity; signal peptide;
4.0
0.01



transmembrane


CSTF1
One of the multiple factors required for polyadenylation
4.0
0.04



and 3′-end cleavage of mammalian pre-mRNAs. May be



responsible for the interaction of CSTF with other factors



to form a stable complex on the pre-mRNA


KIF1A
Motor for anterograde axonal transport of synaptic
4.0
0.02



vesicle precursors


Q96T82
signal peptide; transmembrane
4.0
0.00


ARID3A
Binds a VH promoter proximal site necessary for
3.9
0.02



induced mu-heavy-chain transcription.


CCL7
Chemotactic factor that attracts monocytes and
3.9
0.00



eosinophils, but not neutrophils. This protein can bind



heparin. Binds to CCR1, CCR2 and CCR3


FTCD
Folate-dependent enzyme, that displays both transferase
3.8
0.03



and deaminase activity. Serves to channel one-carbon



units from formiminoglutamate to the folate pool


LRRN1
Cysteine-rich flanking region, C-terminal; Fibronectin,
3.8
0.03



type III; Immunoglobulin-like; Leucine-rich repeat


PTGS2
May have a role as a major mediator of inflammation
3.8
0.03



and/or a role for prostanoid signaling in activity-



dependent plasticity


MATP
Melanocyte differentiation antigen. May transport
3.8
0.06



substances required for melanin biosynthesis


Q7Z5V3
Latrophilin receptor; Olfactomedin-like
3.8
0.03


Q8NG51
Zn-finger, Ran-binding
3.8
0.01


Q86XN7
Aldehyde dehydrogenase; Proline-rich extensin
3.8
0.01


ONECUT2
Transcriptional activator. Activates the transcription of a
3.8
0.02



number of liver genes such as HNF3B


GNS
N-acetylglucosamine-6-sulfatase precursor;
3.7
0.04


COLEC12
Protein C2orf4; C21orf19-like protein
3.7
0.01


IL1A
Produced by activated macrophages, IL-1α stimulates
3.7
0.00



thymocyte proliferation by inducing IL-2 release, B-cell



maturation and proliferation, and fibroblast growth factor



activity. IL-1 proteins are involved in the inflammatory



response, being identified as endogenous pyrogens, and



are reported to stimulate the release of prostaglandin and



collagenase from synovial cells


TNFRSF9
Receptor for TNFSF14/4-1BBL. Possibly active during
3.7
0.02



T cell activation


SLC16A10
T-type amino acid transporter 1; solute carrier family 16,
3.7
0.04



# 10


Q9H9V9
Transcription factor jumonji, jmjC
3.7
0.01


Q9NXL6
SID1 transmembrane family, member 1
3.7
0.04


Q9NTI6
low complexity
3.7
0.03


ASTN2
Fibronectin, type III
3.7
0.06


DAZAP1
Proline-rich extensin; RNA-binding region RNP-1
3.7
0.04


RBM3
Putative RNA-binding protein 3;
3.6
0.02


TEX14
Ankyrin; Protein kinase
3.6
0.04


Q9H631
Mak10 subunit, NatC N(alpha)-terminal
3.6
0.05



acetyltransferase


C16orf3
Protein C16orf3
3.6
0.06


HYAL4
EGF-like domain; Glycoside hydrolase, family 56;
3.6
0.03



sperm surface protein PH20; Multicopper oxidase, type 1


TACSTD1
GA733 tumor-associated antigen gene family may
3.6
0.04



function as growth factor receptors


PLA1A
Esterase/lipase/thioesterase, active site;
3.6
0.01


Q8IZ41;
ATP/GTP-binding site motif A (P-loop); Calcium-
3.5
0.02


Q96N04
binding EF-hand; Ras GTPase superfamily


BACH1
Transcriptional regulator that acts as repressor or
3.5
0.03



activator. Binds, in-vitro, to NF-E2 binding sites. Play



important roles in coordinating transcription activation



and repression by MAFK


SULT1C1
Catalyzes the sulfate conjugation of many drugs,
3.5
0.02



xenobiotic compounds, hormones, and neurotransmitters.


KPNB1
Functions in nuclear protein import, either in association
3.5
0.05



with an adapter protein, like an importin-alpha subunit,



which binds to nuclear localization signals (NLS) in



cargo substrates, or by acting as autonomous nuclear



transport receptor. Acting autonomously, serves itself as



NLS receptor.


CCR7
Receptor for the MIP-3β chemokine. Probable mediator
3.5
0.02



of EBV effects on B lymphocytes or of normal



lymphocyte functions


Q8NC34
Immunoglobulin-like
3.5
0.04


GSK3B
Participates in the Wnt signaling pathway. Implicated in
3.5
0.03



the hormonal control of several regulatory proteins



including glycogen synthase, MYB and the transcription



factor JUN. Phosphorylates JUN at sites proximal to its



DNA-binding domain, thereby reducing its affinity for



DNA


ACSL6
Activation of long-chain fatty acids for both synthesis of
3.5
0.03



cellular lipids, and degradation via beta-oxidation.


H2NC000011
unknown
3.5
0.04


KLRB1
C-type lectin
3.5
0.03


GPR30
Orphan receptor; possibly for a chemokine
3.4
0.02


TNIP3
Listeria induced gene; TNFAIP3 interacting protein 3
3.4
0.02


DBI
Binds medium- and long-chain acyl-CoA esters with
3.4
0.03



very high affinity and may function as an intracellular



carrier of acyl-CoA esters.


PIP5K1B
Phosphatidylinositol-4-phosphate 5-kinase
3.4
0.03


Q92519
Protein kinase
3.4
0.02


PKIG
Extremely potent competitive inhibitor of cAMP-
3.4
0.03



dependent protein kinase activity, this protein interacts



with the catalytic subunit of the enzyme after the cAMP-



induced dissociation of its regulatory chains


STMN2
May play a role in neuronal differentiation, and in
3.3
0.03



modulating membrane interaction with the cytoskeleton



during neurite outgrowth


NCK1
Adapter protein which associates with tyrosine-
3.3
0.04



phosphorylated growth factor receptors or their cellular



substrates


ZFYVE20
Zn-finger, C2H2 type, FYVE type
3.3
0.03


ATP2B1
This magnesium-dependent enzyme catalyzes the
3.3
0.00



hydrolysis of ATP coupled with the transport of calcium



out of the cell


Q96PN6
ATP/GTP-binding site motif A (P-loop); Guanylate
3.3
0.03



cyclase


SOD2
Destroys radicals which are normally produced within
3.3
0.01



the cells and which are toxic to biological systems


VBP1
Binds specifically to cytosolic chaperonin (c-CPN) and
3.3
0.02



transfers target proteins to it. Binds to nascent



polypeptide chain and promotes folding.


CXCL2
Produced by activated monocytes and neutrophils and
3.3
0.01



expressed at sites of inflammation. Hematoregulatory



chemokine, which, in vitro, suppresses hematopoietic



progenitor cell proliferation.


MADH7
Antagonist of signaling by TGFβ (Transforming growth
3.2
0.05



factor) type 1 receptor superfamily members; has been



shown to inhibit TGFβ (Transforming growth factor) and



activin signaling by associating with their receptors thus



preventing SMAD2 access. Functions as an adaptor to



recruit SMURF2 to the TGFβ receptor complex.



SMAD7 is an inhibitory SMAD (I-SMAD) or



antagonistic SMAD whose inhibitory activity is



enhanced by SMURF2


C21orf127
Putative N6-DNA-methyltransferase; M.HsaHemK2P
3.2
0.04


CCDC5
Coiled-coil domain containing 5 (spindle associated).
3.2
0.01


GCH1
Isoform GCH-1 is the functional enzyme, enzymatically
3.2
0.04



inactive isoforms may have other functions


BPAG1
Cytoskeletal linker protein. Anchors keratin-containing
3.2
0.05



intermediate filaments to the inner plaque of



hemidesmosomes. The proteins may self-aggregate to



form filaments or a two-dimensional mesh


DVL2
May play a role in the signal transduction pathway
3.1
0.06



mediated by multiple Wnt genes


GMEB2
Trans-acting factor that binds to glucocorticoid
3.1
0.05



modulatory elements (GME) present in the TAT



(tyrosine aminotransferase) promoter and increases



sensitivity to low concentrations of glucocorticoids.


MARK3
Involved in the specific phosphorylation of microtubule-
3.1
0.02



associated proteins for tau, MAP2 and MAP4.



Phosphorylates CDC25C on Ser-216


PHLDA2
Pleckstrin-like
3.1
0.01


HERC2
Cytochrome b5; Protein kinase; Regulator of
3.1
0.02



chromosome condensation, RCC1; Zn-finger, ZZ type


PTS
Involved in the biosynthesis of tetrahydrobiopterin, an
3.1
0.00



essential cofactor of aromatic amino acid hydroxylases.


HEY2
Antifreeze protein, type I; Basic helix-loop-helix
3.0
0.03



dimerization domain bHLH


NPR1
Receptor for atrial natriuretic peptide. Has guanylate
3.0
0.03



cyclase activity on binding of ANF


FMR2
AF4/FMR2 family member 2; Fragile X mental
3.0
0.02



retardation 2 syndrome protein; Ox19 protein;


Q8N958
unknown
3.0
0.00


NEF3
Neurofilaments usually contain three intermediate
3.0
0.00



filament proteins: L, M, and H which are involved in the



maintenance of neuronal caliber


SNTB2
Adapter protein that binds to and probably organizes the
3.0
0.02



subcellular localization of a variety of membrane



proteins. May link various receptors to the actin



cytoskeleton and the dystrophin glycoprotein complex.



May play a role in the regulation of secretory granules



via its interaction with PTPRN


SOD3
Destroys radicals which are normally produced within
3.0
0.03



the cells and which are toxic to biological systems


C21orf42
Protein C21orf42
3.0
0.03


EREG
May be a mediator of localized cell proliferation. As a
2.9
0.01



mitogen it may stimulate cell proliferation and/or



angiogenesis


OR1F2
Putative odorant receptor
2.9
0.02


Q96HQ3
low complexity
2.9
0.03


CCL2
Chemotactic factor that attracts monocytes and basophils
2.9
0.01



but not neutrophils or eosinophils. Has been implicated



in the pathogenesis of diseases characterized by



monocytic infiltrates, like psoriasis, rheumatoid arthritis



or atherosclerosis.


METTL4
Bipartite nuclear localization signal; MT-A70; N-6
2.9
0.03



Adenine-specific DNA methylase


O60290
KRAB box
2.9
0.04


PLK4
Protein kinase; Tyrosine protein kinase
2.8
0.06


COX7B
One of the polypeptide chains of cytochrome c oxidase,
2.8
0.01



the terminal oxidase in mitochondrial electron transport


GNG2
Guanine nucleotide-binding proteins (G proteins) are
2.8
0.02



involved as modulators or transducers in various



transmembrane signaling systems. The beta and gamma



chains are required for the GTPase activity, for



replacement of GDP by GTP, and for G protein-effector



interaction


MTRF1
Mitochondrial peptide chain release factor that directs
2.8
0.04



the termination of translation in response to the peptide



chain termination codons UAA and UAG


X59362.1
Phospholipid hydroperoxide glutathione peroxidase,
2.8
0.05



mitochondrial precursor (EC 1.11.1.12) (PHGPx) (GPX-



4).


Q96SU5
low complexity; transmembrane
2.8
0.03


NFKB2
NFκB subunits p52 and p100 are respectively the minor
2.8
0.01



and major forms. Appear to have dual functions such as



cytoplasmic retention of attached NFκB proteins and



generation of p52 by a cotranslational processing. The



proteasome-mediated process ensures the production of



both p52 and p100 and preserves their independent



function. p52 binds to the kappa-B consensus sequence



5′-GGRNNYYCC-3′, located in the enhancer region of



genes involved in immune response and acute phase



reactions.


EBF3
Transcriptional activator which recognizes variations of
2.8
0.05



the palindromic sequence 5′-ATTCCCNNGGGAATT-



3′; Transcriptional factor which recognizes variations of



the palindromic sequence 5′-ATTCCCNNGGGAATT-



3′;


Q9H5A9
60S Acidic ribosomal protein
2.8
0.00


NICE1
NICE-1 protein
2.8
0.00


ADRM1
Promotes cell adhesion
2.8
0.00


ATP2B4
This magnesium-dependent enzyme catalyzes the
2.8
0.05



hydrolysis of ATP coupled with the transport of calcium



out of the cell


Q86V45
G-protein beta WD-40 repeat
2.8
0.03


REG1A
Might act as an inhibitor of spontaneous calcium
2.8
0.05



carbonate precipitation. May be associated with neuronal



sprouting in brain, and with brain and pancreas



regeneration


PLAUR
Acts as a receptor for urokinase plasminogen activator.
2.8
0.03



Plays a role in localizing and promoting plasmin



formation. Mediates the proteolysis-independent signal



transduction activation effects of U-PA.


PSMA1
Proteasome subunit, a multicatalytic proteinase complex
2.7
0.04



with ATP-dependent proteolytic activity.


G0S2
Potential oncogene and regulator of latent HIV
2.7
0.03


ITPKB
Inositol 1,4,5-trisphosphate 3-kinase B; IP3K-B
2.7
0.04


PWP2H
Periodic tryptophan protein 2 homolog
2.7
0.04


Q9H8U7
coiled-coil; low complexity
2.7
0.02


Q96BW9
unknown
2.7
0.05


ZNF595
KRAB box; Zn-finger, C2H2 subtype
2.7
0.06


MLLT3
Protein AF-9; Myeloid/lymphoid or mixed-lineage
2.7
0.01



leukemia translocated to chromosome 3 protein


PMSCL1
Component of the nuclear exosome exoribonuclease
2.7
0.04



complex. Required for the 3′ processing of the 7S pre-



RNA to the mature 5.8S rRNA. Has a 3‘5’ exonuclease



activity


UBCE7IP1
Isoform 1 acts as an E3 ubiquitin ligase. Promotes
2.7
0.06



degradation of TLR4 amd TLR9. Isoform 3/ZIN inhibits



TNF and IL-1 mediated activation of NFκB. Promotes



TNF and RIP mediated apoptosis.


CTLA4
Possibly involved in T-cell activation. Binds to B7-1
2.7
0.01



(CD80) and B7-2 (CD86)


Q8N2S5
Pistil-specific extensin-like protein; Proline-rich extensin
2.7
0.05


Q96EC8
Protein of unknown function DUF649
2.7
0.05


YWHAH
Adapter protein implicated in the regulation of a large
2.6
0.03



spectrum of both general and specialized signaling



pathways. Binds to a large number of partners, usually



by recognition of a phosphoserine or phosphothreonine



motif. Binding generally results in the modulation of the



activity of the binding partner


MGAT5
Catalyzes the addition of N-acetylglucosamine in beta 1-
2.6
0.01



6 linkage to the alpha-linked mannose of biantennary N-



linked oligosaccharides. It is one of the most important



enzymes involved in the regulation of the biosynthesis of



glycoprotein oligosaccharides


DNAH5
ATP/GTP-binding site motif A (P-loop); Dynein heavy
2.6
0.04



chain; Eukaryotic thiol (cysteine) protease


SPG7
2Fe—2S ferredoxin; Peptidase M41
2.6
0.02


INSIG1
May play a role in growth and differentiation of tissues
2.6
0.02



involved in metabolic control. May play a regulatory role



during G0/G1 transition of cell growth


PANK3
Plays a role in the physiological regulation of the
2.6
0.02



intracellular CoA concentration


Q8WUC7
Bipartite nuclear localization signal
2.6
0.00


HDC
Histidine decarboxylase; HDC
2.6
0.04


DGKA
Upon cell stimulation converts the second messenger
2.6
0.03



diacylglycerol into phosphatidate, initiating the



resynthesis of phosphatidylinositols and attenuating



protein kinase C activity


HNF4A
Transcriptionally controlled transcription factor. Binds to
2.6
0.01



DNA sites required for the transcription of alpha 1-



antitrypsin, apolipoprotein CIII, transthyretin genes and



HNF1-alpha.


MAP1LC3A
Probably involved in formation of autophagosomal
2.5
0.04



vacuoles


DACH2
Bipartite nuclear localization signal; Transforming
2.5
0.04



protein Ski


ZNF80
May be involved in transcriptional regulation
2.5
0.02


TNFAIP6
Possibly involved in cell-cell and cell-matrix interactions
2.5
0.00



during inflammation and tumorigenesis


HIF1A
Functions as a master transcriptional regulator of the
2.5
0.04



adaptive response to hypoxia. Under hypoxic conditions



activates the transcription of over 40 genes, including,



erythropoietin, glucose transporters, glycolytic enzymes,



vascular endothelial growth factor, and other genes



whose protein products increase oxygen delivery or



facilitate metabolic adaptation to hypoxia. Binds to core



DNA sequence 5′-[AG]CGTG-3′; within the hypoxia



response element (HRE) of target gene promoters.



Activation requires recruitment of transcriptional



coactivators such as CREBPB and EP300. Activity is



enhanced by interaction with both, NCOA1 or NCOA2.



Interaction with redox regulatory protein APEX seems to



activate CTAD and potentiates activation by NCOA1



and CREBBP


SLC39A8
Zinc transporter ZIP
2.5
0.01


MAFF
Interacts with the upstream promoter region of the
2.5
0.00



oxytocin receptor gene. May be involved in the cellular



stress response


CD22
Mediates B-cell B-cell interactions. Binds sialylated
2.5
0.06



glycoproteins; one of which is CD45. Preferentially



binds to alpha2,6-linked sialic acid. Upon ligand induced



tyrosine phosphorylation in the immune response seems



to be involved in regulation of B cell antigen receptor



signaling. Plays a role in positive regulation through



interaction with Src family tyrosine kinases and may also



act as an inhibitory receptor by recruiting cytoplasmic



phosphatases via their SH2 domains that block signal



transduction through dephosphorylation of signaling



molecules


S100A12
Calcitermin possesses antifungal activity against
2.5
0.03




C. albicans and is also active against E. coli and




P. aeruginosa but not L. monocytogenes and S. aureus


IL1F9
Function as an agonist of NFκ B activation through the
2.5
0.03



orphan IL-1-receptor-related protein 2. Could constitute



part of an independent signaling system analogous to



interleukin-1α, β receptor agonist and interleukin-1



receptor type I (IL-1R1), that is present in epithelial



barriers and takes part in local inflammatory response


Q9Y3U6
low complexity
2.5
0.00


SERPINB8
Serpin B8; Cytoplasmic antiproteinase 2; CAP2; CAP-2;
2.5
0.03



Protease inhibitor 8


CEBPD
C/EBP is a DNA-binding protein that recognizes two
2.5
0.05



different motifs: the CCAAT homology common to



many promoters and the enhanced core homology



common to many enhancers. Important transcriptional



activator in the regulation of genes involved in immune



and inflammatory responses, may play an important role



in the regulation of the several genes associated with



activation and/or differentiation of macrophages


ATP13A3
Probable cation-transporting ATPase 13A3; ATPase
2.5
0.03



family homolog up-regulated in senescence cells 1


NAP1L3
Nucleosome assembly protein 1-like 3
2.5
0.01


KIAA1404
Protein KIAA1404
2.5
0.04


C6orf103
IQ calmodulin-binding region
2.5
0.01


Q96DM7
Cytochrome b5
2.4
0.04


ID2
ID (inhibitor of DNA binding) HLH proteins lack a basic
2.4
0.00



DNA-binding domain but are able to form heterodimers



with other HLH proteins, thereby inhibiting DNA



binding. ID-2 may be an inhibitor of tissue-specific gene



expression


BGLAP
Constitutes 1-2% of the total bone protein. It binds
2.4
0.06



strongly to apatite and calcium


AQP9
Forms a channel with a broad specificity, mediates
2.4
0.00



passage of a wide variety of non-charged solutes


C5orf13
Neuronal protein 3.1; p311 protein
2.4
0.01


PROZ
Appears to assist hemostasis by binding thrombin and
2.4
0.05



promoting its association with phospholipid vesicles


COL3A1
Collagen type III occurs in most soft connective tissues
2.4
0.00



along with type I collagen


TNF
Tumour necrosis factor-α; Cytokine that binds to
2.4
0.01



TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR. It is



mainly secreted by macrophages and can induce cell



death of certain tumor cell lines. It is potent pyrogen



causing fever by direct action or by stimulation of



interleukin 1 secretion and is implicated in the induction



of cachexia, Under certain conditions it can stimulate



cell proliferation and induce cell differentiation


CD151
Essential for the proper assembly of the glomerular and
2.4
0.02



tubular basement membranes in kidney


RAB3B
Protein transport. Probably involved in vesicular traffic
2.4
0.00


TUSC4
Bipartite nuclear localization signal
2.4
0.01


PRDM2
May function as a DNA-binding transcription factor.
2.4
0.03



Binds to the macrophage-specific TPA-responsive



element (MTE) of the HMOX1 (heme oxygenase 1) gene



and may act as a transcriptional activator of this gene


DEF6
Calcium-binding EF-hand; Pleckstrin-like; Tropomyosin
2.4
0.02


Q9H864
Bipartite nuclear localization signal
2.4
0.05


HRAS
Ras proteins bind GDP/GTP and possess intrinsic
2.4
0.01



GTPase activity


SPTBN1
Fodrin, which seems to be involved in secretion,
2.4
0.05



interacts with calmodulin in a calcium-dependent manner



and is thus candidate for the calcium-dependent



movement of the cytoskeleton at the membrane


ETV5
Binds to DNA sequences containing the consensus
2.4
0.01



nucleotide core sequence GGAA


C7orf16
Inhibits protein phosphatase-2A and protein
2.4
0.00



phosphatase-1


Q96IB9
low complexity
2.4
0.04


ODZ1
EGF-like domain; Laminin-type EGF-like domain; NHL
2.3
0.00



repeat; Proline-rich region; Subtilase serine protease


OR7A5
Putative odorant receptor
2.3
0.00


FIGNL1
AAA ATPase, central region; AAA-protein subdomain;
2.3
0.00



ATP/GTP-binding site motif A (P-loop)


Q9Y3B9
Bipartite nuclear localization signal
2.3
0.04


SRPRB
Has GTPase activity. May mediate the membrane
2.3
0.05



association of SR alpha


CCNB1IP1
E3 ubiquitin ligase. Modulates cyclin B levels and
2.3
0.01



participates in the regulation of cell cycle progression



through the G2 phase. Overexpression causes delayed



entry into mitosis


DSG3
Component of intercellular desmosome junctions.
2.3
0.02



Involved in the interaction of plaque proteins and



intermediate filaments mediating cell-cell adhesion


C2orf6
Mob1/phocein family
2.3
0.03


LILRB1
Receptor for class I MHC antigens. Recognizes a broad
2.3
0.00



spectrum of HLA-A, HLA-B, HLA-C and HLA-G



alleles. Ligand binding results in inhibitory signals and



down-regulation of the immune response. Engagement



of LILRB1 present on natural killer cells or T-cells by



class I MHC molecules protects the target cells from



lysis.


O14950
Calcium-binding EF-hand
2.3
0.04


ZNF576
Zn-finger, C2H2 type
2.3
0.02


WTAP
Wilms' tumor 1-associating protein; Putative pre-mRNA
2.3
0.01



splicing regulator female-lethal(2D) homolog


PTP4A3
Prenyl group binding site (CAAX box); Tyrosine
2.3
0.03



specific protein phosphatase and dual specificity protein



phosphatase


ELF1
Transcription factor that appears to be required for the T-
2.3
0.03



cell-receptor-mediated trans activation of HIV-2 gene



expression. Activates the LYN and mouse BLK



promoters


GDA
Catalyzes the hydrolytic deamination of guanine,
2.3
0.01



producing xanthine and ammonia


IL6
IL-6 is a cytokine with a wide variety of biological
2.3
0.03



functions: it plays an essential role in the final



differentiation of B-cells into Ig-secreting cells, it



induces myeloma and plasmacytoma growth, it induces



nerve cells differentiation, in hepatocytes it induces acute



phase reactants


Q9BWJ2
unknown
2.3
0.03


Q8TF23
BED finger; Cytochrome c heme-binding site; KRAB
2.3
0.02



box; Zn-finger, C2H2 subtype


APTX
Histidine triad (HIT) protein; Zn-finger, C2H2 type
2.3
0.03


Q96LP3
Leucine-rich repeat
2.3
0.02


MAB21L1
Mab-21 protein
2.3
0.01


ATP1B1
ATPase B subunit; This is the non-catalytic component
2.3
0.00



of the active enzyme, which catalyzes the hydrolysis of



ATP coupled with the exchange of Na(+) and K(+) ions



across the plasma membrane. The beta subunit regulates,



through assembly of alpha/beta heterodimers, the



number of sodium pumps transported to the plasma



membrane


Q9H095
IQ calmodulin-binding region
2.3
0.04


ENSG00000110900
CD9/CD37/CD63 antigen
2.3
0.02


Q86X05
Hly-III related proteins
2.3
0.00


IBRDC2
Zn-finger, RING; Zn-finger, cysteine-rich C6HC
2.3
0.01


PAPOLG
Responsible for the post-transcriptional adenylation of
2.3
0.02



the 3′-terminal several small RNAs including signal



recognition particle (SRP) RNA, nuclear 7SK RNA, U2



small nuclear RNA, and ribosomal 5S RNA


Q7Z5X7
low complexity
2.2
0.02


EIF3S12
Binds to the 40S ribosome and promotes the binding of
2.2
0.01



methionyl-tRNAi and mRNA


MDN1
May function as a nuclear chaperone in the assembly/
2.2
0.03



disassembly of macromolecular complexes in the



nucleus


C6orf123
Protein C6orf123; HGC6.2 protein
2.2
0.03


ADM
AM and PAMP are potent hypotensive and vasodilatator
2.2
0.01



agents. Numerous actions have been reported most



related to the physiologic control of fluid and electrolyte



homeostasis.


PLAC8
Placenta-specific gene 8 protein; C15 protein
2.2
0.05


CYP19A1
Catalyzes the formation of aromatic C18 estrogens from
2.2
0.03



C19 androgens


ENSG00000144872
Ribosomal protein L39e
2.2
0.00


PPP1CB
Protein phosphatase (PP1) is essential for cell division, it
2.2
0.02



participates in the regulation of glycogen metabolism,



muscle contractility and protein synthesis. Involved in



regulation of ionic conductances and long-term synaptic



plasticity


TRIM36
Butyrophylin-like; Fibronectin, type III; SPla/RYanodine
2.2
0.05



receptor SPRY; Zn-finger, B-box, RING


TRPC7
Thought to form a receptor-activated non-selective
2.2
0.02



calcium permeant cation channel. Probably is operated



by a phosphatidylinositol second messenger system



activated by receptor tyrosine kinases or G-protein



coupled receptors. Activated by diacylglycerol (DAG)



(By similarity). May also be activated by intracellular



calcium store depletion


Q15061
G-protein beta WD-40 repeat
2.2
0.03


CACNA1H
Voltage-sensitive calcium channels (VSCC) mediate the
2.2
0.01



entry of calcium ions into excitable cells and are also



involved in a variety of calcium-dependent processes,



including muscle contraction, hormone or



neurotransmitter release, gene expression, cell motility,



cell division and cell death. processing as well as in cell



growth processes


Q8N9J0
FUN14 family
2.2
0.01


TNFAIP8
Tumor necrosis factor, alpha-induced protein 8
2.2
0.05


C1orf24
Niban protein
2.2
0.04


DUSP5
Displays phosphatase activity toward several substrates.
2.2
0.04



The highest relative activity is toward ERK1


AP4B1
Subunit of novel type of clathrin-or non-clathrin-
2.2
0.05



associated protein coat involved in targeting proteins



from the trans-Golgi network (TGN) to the endosomal-



lysosomal system


Q96MN5
unknown
2.2
0.03


EBI3
Cytokine receptor, common beta/gamma chain;
2.2
0.02



Fibronectin, type III; Long hematopoietin receptor,



soluble alpha chain


PELI1
Scaffold protein involved in the IL-1 signaling pathway
2.2
0.01



via its interaction with the complex containing IRAK



kinases and TRAF6. Required for NF-kappa-B activation



and IL-8 gene expression in response to IL-1


PRPF3
Participates in pre-mRNA splicing. May play a role in
2.2
0.03



the assembly of the U4/U5/U6 tri-snRNP complex


SLC7A13
Amino acid permease-associated region; Amino acid/
2.2
0.04



polyamine transporter, family I


MAP2K2
Catalyzes the concomitant phosphorylation of a
2.2
0.03



threonine and a tyrosine residue in a Thr-Glu-Tyr



sequence located in MAP kinases. Activates the ERK1



and ERK2 MAP kinases


DDX21
Can unwind double-stranded RNA (helicase) and can
2.2
0.00



fold or introduce a secondary structure to a single-



stranded RNA (foldase). Functions as cofactor for c-Jun-



activated transcription. Involved in rRNA processing


GCN5L1
Biogenesis of lysosome-related organelles complex-1,
2.2
0.01



subunit 1; BLOC-1 subunit 1; GCN5-like protein 1;



RT14 protein


SERPINB8
Inhibits urokinase-type plasminogen activator.
2.2
0.02


Q9UJA5
Bipartite nuclear localization signal; Eukaryotic
2.2
0.04



initiation factor 3, gamma subunit


CD81
May play an important role in the regulation of
2.1
0.00



lymphoma cell growth. Interacts with a 16-kDa Leu-13



protein to form a complex possibly involved in signal



transduction.


BID
Induces ICE-like proteases and apoptosis. Counters the
2.1
0.06



protective effect of Bcl-2


OR8B8
Putative odorant receptor; Putative odorant receptor.
2.1
0.04



Could also be involved in taste perception


RBMS3
Paraneoplastic encephalomyelitis antigen; RNA-binding
2.1
0.00



region RNP-1 (RNA recognition motif)


UNC5B
ATP/GTP-binding site motif A (P-loop); Death domain;
2.1
0.04



Immunoglobulin-like; Thrombospondin, type I; ZU5



domain


ENSG00000154511
low complexity; transmembrane
2.1
0.00


ERCC6
Is involved in the preferential repair of active genes.
2.1
0.01



Presumed DNA or RNA unwinding function.


PL6
PL6 protein; Placental protein 6; PP6
2.1
0.05


IGHG3
Ig alpha is the major immunoglobulin class in body
2.1
0.00



secretions. It serves both to defend against local infection



and to prevent access of foreign antigens to the general



immunologic system


RIPK2
Activates pro-caspase-1 and pro-caspase-8. Potentiates
2.1
0.02



CASP-8-mediated apoptosis. Activates NFκB


HDAC4
Responsible for the deacetylation of lysine residues on
2.1
0.01



the N-terminal part of the core histones (H2A, H2B, H3



and H4). Histone deacetylation gives a tag for epigenetic



repression and plays an important role in transcriptional



regulation, cell cycle progression and developmental



events.


SOD2
Destroys radicals which are normally produced within
2.1
0.00



the cells and which are toxic to biological systems


DDX39
ATP/GTP-binding site motif A; DEAD/DEAH box
2.1
0.03



helicase


ADORA2A
Receptor for adenosine. The activity of this receptor is
2.1
0.03



mediated by G proteins which activate adenylyl cyclase


O15069
Nascent polypeptide-associated complex NAC
2.1
0.04


CPD
Metallocarboxypeptidase D precursor, gp180
2.1
0.03


Q9P233
Calponin-like actin-binding; Eggshell protein; Leucine-
2.1
0.03



rich repeat


TTLL3
Tubulin tyrosine ligase-like protein 3; HOTTL
2.1
0.05


C16orf44
BTB/POZ domain; Kelch repeat
2.1
0.02


Q9Y627
Aldehyde dehydrogenase; Leucine-rich repeat
2.1
0.03


OAZIN
Inhibits antizyme-dependent ornithine decarboxylase
2.1
0.03



degradation by binding to antizyme


RRBP1
Acts as a ribosome receptor and mediates interaction
2.1
0.03



between the ribosome and the endoplasmic reticulum



membrane


BOK
Apoptosis regulator Bcl-2 protein, BH; BCL2-like
2.1
0.03



apoptosis inhibitor


RANBP9
Proline-rich extensin; Proline-rich region;
2.1
0.03



SPla/RYanodine receptor SPRY


IL7R
Receptor for interleukin-7
2.1
0.02


Q9NW83
low complexity
2.0
0.04


FER1L3
May play a role in membrane regeneration and repair
2.0
0.03


CD44
Receptor for hyaluronic acid (HA). Mediates cell-cell
2.0
0.01



and cell-matrix interactions. Adhesion with HA plays an



important role in cell migration, tumor growth and



progression. Also involved in lymphocyte activation,



recirculation and homing, and in hematopoiesis.


ENSG00000187017
ATP/GTP-binding site motif A (P-loop); Actin-binding
2.0
0.06



WH2; Bipartite nuclear localization signal


Q8IVG4
low complexity
2.0
0.01


KIAA0084
Hypothetical protein KIAA0084; HA2022
2.0
0.04


Q9UF01
FGF receptor activating protein 1
2.0
0.01


NEFH
Neurofilaments usually contain three intermediate
2.0
0.03



filament proteins: L, M, and H which are involved in the



maintenance of neuronal caliber. NF-H has an important



function in mature axons that is not subserved by the two



smaller NF proteins


FNDC5
Fibronectin, type III
2.0
0.00


KIAA1533
GRAM domain
2.0
0.01


ZWINT
May play a role in targeting HZW10 to the kinetochore
2.0
0.06



at prometaphase. Part of the MIS12 complex, which may



be fundamental for kinetochore formation and proper



chromosome segregation during mitosis


PNRC1
Nuclear receptor coactivator. May play a role in signal
2.0
0.05



transduction


SLAMF7
SLAM family member; 719A24. protein; CD2-like
2.0
0.04



receptor activating cytotoxic cells.


KLK1
Glandular kallikreins cleave Met-Lys and Arg-Ser bonds
2.0
0.00



in kininogen to release Lys-bradykinin


Q9P0P9
transmembrane
2.0
0.01


NEK1
Phosphorylates serines and threonines, but also appears
2.0
0.04



to have tyrosine kinase activity. Implicated in control of



meiosis


Q9H6X1
Proline-rich extensin; Proline-rich region
2.0
0.03


ARIH1
Might act as an E3 ubiquitin-protein ligase, or as part of
2.0
0.06



the E3 complex, which accepts ubiquitin from specific



E2 ubiquitin-conjugating enzymes, such as



UBE2L3/UBCM4, and then transfers it to substrates


ZFP90
May function as a repressor or silencer protein, and most
2.0
0.03



likely exerts its repressing activity upon zinc-dependent



binding to DNA. May be involved in proper



spermatogenesis by repressing the expression of genes



unnecessary or incompatible with the maintenance of a



haploid cell state


TRIM39
Tripartite motif protein 39; RING finger protein 23;
2.0
0.02



Testis-abundant finger protein


Q96NU6
Bipartite nuclear localization signal; RhoGAP domain
2.0
0.01


IRAK3
Death domain; Protein kinase
2.0
0.03


Q9NTF2
Prenyl group binding site (CAAX box)
2.0
0.03


PIP5K3
Supports the intracellular PIP pool and to a lesser extent,
2.0
0.00



the PI 4,5-P(2) pool. It generates PIP from PI and, to a



lesser extent, PI 4,5-P(2) from PI 4-P. There are



indications that it phosphorylates the D-5 rather than the



D-4 position. Has a role in endosome-related membrane



trafficking


EIF3S7
Binds to the 40S ribosome and promotes the binding of
2.0
0.05



methionyl-tRNAi and mRNA. Associates with the



subunit p170 of eIF-3


NME1
Major role in the synthesis of nucleoside triphosphates
2.0
0.00



other than ATP


GDAP1L1
Ganglioside-induced differentiation-associated protein 1-
2.0
0.02



like 1; GDAP1-L1


Q8NBH1
unknown
2.0
0.00


TCF12
Binds specifically to oligomers of E-box motifs. May
2.0
0.03



play important roles during development of the nervous



system as well as in other organ systems


Q8TEB0
unknown
2.0
0.04


LY6G5C
C-type lectin
2.0
0.01


ZNF451
May be involved in transcriptional regulation.
2.0
0.03



Coactivator for steroid receptors


PLEKHF2
Pleckstrin-like; Zn-finger, FYVE type
2.0
0.04


NDUFB4
Transfer of electrons from NADH to the respiratory
2.0
0.02



chain. The immediate electron acceptor for the enzyme is



believed to be ubiquinone


ATCAY
Caytaxin; Ataxia Cayman type protein; BNIP-H
2.0
0.01


GP5
The GPIb-V-IX complex functions as the von
2.0
0.05



Willebrand factor receptor and mediates von Willebrand



factor-dependent platelet adhesion to blood vessels. The



adhesion of platelets to injured vascular surfaces in the



arterial circulation is a critical initiating event in



hemostasis


ETV4
Ets-domain; PEA3-type ETS-domain transcription
2.0
0.05



factor, N-terminal


TNFAIP3
Interacts with NAF1 and inhibits TNF-induced NF-
2.0
0.04



kappa-B-dependent gene expression by interfering with



an RIP- or TRAF2-mediated transactivation signal.



Inhibitor of programmed cell death. Has a role in the



function of the lymphoid system and may contribute to



the in vivo effects of TNFα. Has deubiquitinating



activity that is directed towards Lys-48 or Lys-63-linked



polyubiquitin chains


OASL
Does not have 2′;-5′-OAS activity, but binds double-
2.0
0.01



stranded RNA and DNA


HBG1
The epsilon chain is a beta-type chain of early
2.0
0.04



mammalian embryonic hemoglobin


PAPOLB
Polymerase that creates the 3′ poly(A) tail of
2.0
0.00



mRNA&apos; s. Also required for the



endoribonucleolytic cleavage reaction at some



polyadenylylation sites. May acquire specificity through



interaction with a cleavage and polyadenylation



specificity factor (CPSF) at its C-terminus


ADA
Adenosine/AMP deaminase; Adenosine/AMP deaminase
1.9
0.03



active site


LAMA2
Binding to cells via a high affinity receptor, laminin is
1.9
0.02



thought to mediate the attachment, migration and



organization of cells into tissues during embryonic



development by interacting with other extracellular



matrix components


F13A1
Factor XIII is activated by thrombin and calcium ion to a
1.9
0.03



transglutaminase that catalyzes the formation of gamma-



glutamyl-epsilon-lysine cross-links between fibrin



chains, thus stabilizing the fibrin clot. Also cross-link



alpha-2-plasmin inhibitor, or fibronectin, to the alpha



chains of fibrin


SIAT4C
It may catalyze the formation of the NeuAc-alpha-2,3-
1.9
0.03



Gal-beta-1,3-GalNAc- or NeuAc-alpha-2,3-Gal-beta-1,3-



GlcNAc-sequences found in terminal carbohydrate



groups of glycoproteins and glycolipids. It may be



involved in the biosynthesis of the sialyl Lewis X



determinant


BTBD12
BTB/POZ domain
1.9
0.06


APOBEC2
Probable C to U editing enzyme whose physiological
1.9
0.02



substrate is not yet known. Does not display detectable



apoB mRNA editing. Has a low intrinsic cytidine



deaminase activity


RIN2
Ras effector protein. May function as an upstream
1.9
0.04



activator and/or downstream effector for RAB5B in



endocytic pathway. May function as a guanine



nucleotide exchange (GEF) of RAB5B, required for



activating the RAB5 proteins by exchanging bound GDP



for free GTP


UFC1
E2-like enzyme which forms an intermediate with UFM1
1.9
0.01



via a thioester linkage


ZNF83
ATP/GTP-binding site motif A (P-loop); Bipartite
1.9
0.01



nuclear localization signal; KRAB box; Zn-finger, C2H2



subtype;


Q86SU1
Ubiquitin interacting motif
1.9
0.05


FRS2
Insulin receptor substrate-1, PTB
1.9
0.04


IER3
Radiation-inducible immediate-early gene IEX-1;
1.9
0.00



Differentiation-dependent gene 2 protein; DIF-2 protein


COX11
Exerts its effect at some terminal stage of cytochrome c
1.9
0.01



oxidase synthesis, probably by being involved in the



insertion of the copper B into subunit I


FBXO32
Probably recognizes and binds to some phosphorylated
1.9
0.02



proteins and promotes their ubiquitination and



degradation during skeletal muscle atrophy


Q96QA0
Mpv17/PMP22
1.9
0.01


GALNT9
Ricin B lectin domain
1.9
0.04


VPREB1
Associates with the Ig-mu chain to form a molecular
1.9
0.05



complex that is expressed on the surface of pre-B-cells



and regulates Ig gene rearrangements in the early steps



of B-cell differentiation


NCAM2
May play important roles in selective fasciculation and
1.9
0.04



zone-to-zone projection of the primary olfactory axons


SLC17A5
General substrate transporter
1.9
0.04


PMM2
Involved in the synthesis of the GDP-mannose and
1.9
0.02



dolichol-phosphate-mannose required for a number of



critical mannosyl transfer reactions


CDC42EP4
Probably involved in the organization of the actin
1.9
0.01



cytoskeleton. May act downstream of CDC42 to induce



actin filament assembly leading to cell shape changes.



Induces pseudopodia formation, when overexpressed in



fibroblasts


HFE
Binds to transferrin receptor (TFR) and reduces its
1.9
0.05



affinity for iron-loaded transferrin


HMG20A
HMG1/2 (high mobility group) box
1.9
0.02


GBP1
Binds GTP, GDP and GMP
1.9
0.01


NOL8
Bipartite nuclear localization signal; RNA-binding
1.9
0.02



region RNP-1 (RNA recognition motif)


C10orf46
Proline-rich region
1.9
0.06


LECT2
Has a neutrophil chemotactic activity. Also a positive
1.9
0.04



regulator of chondrocyte proliferation


Q96136
transmembrane
1.9
0.03


Q9H3H7
Brain my050 protein.
1.9
0.05


DAB1
Adapter molecule functioning in neural development.
1.9
0.01



May regulate SIAH1 activity


ZIC1
May play a role in cerebellar development
1.9
0.02


SIPA1
GTPase activator for the nuclear Ras-related regulatory
1.9
0.04



proteins Rap1 and Rap2 in vitro, converting it to the



putatively inactive GDP-bound state


EHHADH
eIF-2 functions in the early steps of protein synthesis by
1.9
0.02



forming a ternary complex with GTP and initiator tRNA.


NSMAF
Couples the p55 TNF-receptor (TNF-R55/TNFR1) to
1.9
0.01



neutral sphingomyelinase (N-SMASE). Specifically



binds to the N-smase activation domain of TNF-R55.



May regulate ceramide production by N-SMASE


DSPG3
May have a role in bone formation and also in
1.9
0.01



establishing the ordered structure of cartilage through



matrix organization


NR4A3
Binds to the B1A response-element
1.9
0.05


CHRM1
The muscarinic acetylcholine receptor mediates various
1.9
0.03



cellular responses, including inhibition of adenylate



cyclase, breakdown of phosphoinositides and modulation



of potassium channels through the action of G proteins.



Primary transducing effect is PI turnover


Q9Y2F5
low complexity
1.9
0.02


EDG2
Receptor for lysophosphatidic acid (LPA), a mediator of
1.9
0.01



diverse cellular activities. Seems to be coupled to the



G(i)/G(0), G(12)/G(13), and G(q) families of



heteromeric G proteins


Q9HBM0
Plays a pivotal role in the establisment of adherens
1.9
0.01



junctions and their maintenance in adult life


FPR1
High affinity receptor for N-formyl-methionyl peptides,
1.9
0.02



which are powerful neutrophils chemotactic factors.



Binding of FMLP to the receptor causes activation of



neutrophils. This response is mediated via a G-protein



that activates a phosphatidylinositol-calcium second



messenger system


ATF1
This protein binds the cAMP response element (CRE), a
1.9
0.05



sequence present in many viral and cellular promoters.



Binds to the Tax-responsive element (TRE) of HTLV-I.



Mediates PKA-induced stimulation of CRE-reporter



genes


ZCCHC2
Zinc finger CCHC domain containing protein 2
1.9
0.03


MANBAL
Protein MANBAL
1.9
0.01


KIF13A
Plus end-directed microtubule-dependent motor protein
1.9
0.05



involved in mannnose-6-phosphate receptor (M6PR)



transport to the plasma membrane


CYorf15A
Testis protein (Fragment).
1.9
0.01


ARHGDIA
Regulates the GDP/GTP exchange reaction of the Rho
1.9
0.00



proteins by inhibiting the dissociation of GDP from



them, and the subsequent binding of GTP to them


PCDHB10
Potential calcium-dependent cell-adhesion protein. May
1.8
0.01



be involved in the establishment and maintenance of



specific neuronal connections in the brain


TRPC5
Thought to form a receptor-activated non-selective
1.8
0.00



calcium permeant cation channel. Probably is operated



by a phosphatidylinositol second messenger system



activated by receptor tyrosine kinases or G-protein



coupled receptors. May also be activated by intracellular



calcium store depletion


ZDHHC2
Palmitoyltransferase specific for GAP43 and
1.8
0.00



DLG4/PSD95


Q8NEZ3
7-Fold repeat in clathrin and VPS proteins; G-protein
1.8
0.04



beta WD-40 repeat


COQ7
Potential central metabolic regulator
1.8
0.01


DDX3Y
Probable ATP-dependent RNA helicase. May play a role
1.8
0.05



in spermatogenesis


ITPKA
Inositol 1,4,5-trisphosphate 3-kinase A; IP3K-A
1.8
0.02


GDF9
Required for ovarian folliculogenesis
1.8
0.04


ZNF593
Negatively modulates the DNA binding activity of Oct-2
1.8
0.03



and therefore its transcriptional regulatory activity. May



also be a modulator of other octamer-binding proteins


SERTAD1
Acts at E2F-responsive promoters to integrate signals
1.8
0.01



provided by PHD- and/or bromodomain-containing



transcription factors. Stimulates E2F-1/DP-1



transcriptional activity. Renders the activity of cyclin



D1/CDK4 resistant to the inhibitory effects of



p16(INK4a)


SLC22A11
General substrate transporter
1.8
0.04


ZNF578
KRAB box; Zn-finger, C2H2 subtype
1.8
0.03


TROAP
Could be involved with bystin and trophinin in a cell
1.8
0.05



adhesion molecule complex at the time of the embryo



implantation


HAPLN1
Stabilizes the aggregates of proteoglycan monomers with
1.8
0.02



hyaluronic acid in the extracellular cartilage matrix


ETS2
C-ets-2 protein
1.8
0.04


Q96PY3
Leucine-rich repeat
1.8
0.04


DNAJB6
DnaJ homolog subfamily B member 6; Heat shock
1.8
0.05



protein J2;


STAMBP
Bipartite nuclear localization signal; Mov34 family
1.8
0.05


PIP5K2C
Phosphatidylinositol-4-phosphate 5-kinase
1.8
0.03


RNH
Inhibitor of pancreatic RNase and angiogenin. May also
1.8
0.01



function in the modulation of cellular activities


Q8N1W2
Cytochrome c heme-binding site; Zn-finger, C2H2 type
1.8
0.06


Q96AP0
low complexity
1.8
0.01


Q9H6L9
Autophagocytosis associated protein
1.8
0.04


TSGA10
Testis specific, 10.
1.8
0.01


SLC7A5
Sodium-independent, high-affinity transport of large
1.8
0.01



neutral amino acids. Involved in cellular amino acid



uptake


ATR
Phosphatidylinositol 3- and 4-kinase-related, FAT,
1.8
0.01



FATC;


ADRB2
Beta-adrenergic receptors mediate the catecholamine-
1.8
0.05



induced activation of adenylate cyclase through the



action of G proteins.


7NF585A
May be involved in transcriptional regulation
1.8
0.01


MKI67
Thought to be required for maintaining cell proliferation
1.8
0.04


NPAS2
Neuronal PAS domain protein 2; Neuronal PAS2;
1.8
0.03



Member of PAS protein 4; MOP4


WASF3
Downstream effector molecules involved in the
1.8
0.04



transmission of signals from tyrosine kinase receptors



and small GTPases to the actin cytoskeleton


Q96IJ6
Bacterial transferase hexapeptide repeat; Nucleotidyl
1.8
0.02



transferase


Q8NBM8
NULL
1.8
0.02


SIGLEC5
Putative adhesion molecule that mediates sialic-acid
1.8
0.03



dependent binding to cells.


EPHB2
Receptor for members of the ephrin-B family
1.8
0.03


PIK4CB
Phosphorylates phosphatidylinositol (PI) in the first
1.8
0.03



committed step in the production of the second



messenger inositol-1,4,5,-trisphosphate (PIP). May



regulate Golgi disintegration/reorganization during



mitosis, possibly via its phosphorylation


BPAG1
Cytoskeletal linker protein. Anchors keratin-containing
1.8
0.01



intermediate filaments to the inner plaque of hemidesmosomes.



May self-aggregate to form filaments or a 2D



mesh


SERPINB1
Regulates the activity of the neutrophil proteases
1.8
0.03



elastase, cathepsin G and proteinase-3


Q86T73
von Willebrand factor, type A
1.8
0.03


FRMD1
Band 4.1 domain
1.8
0.00


CD79A
Associated to surface IgM-receptor; may be involved in
1.8
0.00



signal transduction


OXA1L
Required for the insertion of integral membrane proteins
1.8
0.05



into the mitochondrial inner membrane. Essential for the



activity and assembly of cytochrome oxidase


HSD3B1
3beta-HSD is a bifunctional enzyme that plays a crucial
1.8
0.02



role in the biosynthesis of all classes of hormonal



steroids


TBL1XR1
F-box-like protein involved in the recruitment of the
1.8
0.00



ubiquitin/19S proteasome complex to nuclear receptor-



regulated transcription units. Plays an essential role in



transcription activation mediated by nuclear receptors.


C21orf124
Required for synthesis of pyridoxal-5-phosphate from
1.8
0.01



vitamin B6


ENSG00000166965
Regulator of chromosome condensation, RCC1
1.8
0.03


PTPN4
May act at junctions between the membrane and the
1.7
0.03



cytoskeleton


ODC1
Ornithine decarboxylase; ODC
1.7
0.05


SLC23A3
Xanthine/uracil/vitamin C permease family
1.7
0.03


KIAA0391
60S ribosomal protein L9; Hypothetical protein
1.7
0.02



KIAA0391


BZW2
ATP/GTP-binding site motif A (P-loop); eIF4-
1.7
0.01



γ/eIF5/eIF2-ε


MAPK6
Phosphorylates microtubule-associated protein 2
1.7
0.01



(MAP2). May promote entry in the cell cycle


RSU1
Potentially plays a role in the Ras signal transduction
1.7
0.01



pathway. Capable of suppressing v-Ras transformation in



vitro


PRDM10
BTB/POZ domain; Zn-finger, C2H2 type
1.7
0.03


SHMT1
Interconversion of serine and glycine
1.7
0.01


Q9Y4C1
Transcription factor jumonji, jmjC
1.7
0.03


RAP1B
Ras-related protein Rap-1b; GTP-binding protein smg
1.7
0.03



p21B


CASP9
Involved in the activation cascade of caspases
1.7
0.02



responsible for apoptosis. Binding of caspase-9 to Apaf-



1 leads to activation of the protease which then cleaves



and activates caspase-3. Proteolytically cleaves



poly(ADP-ribose) polymerase (PARP)


SDPR
Serum deprivation response protein; Phosphatidylserine-
1.7
0.02



binding protein.


RBM8A
Part of a post-splicing multiprotein complex involved in
1.7
0.04



both mRNA nuclear export and mRNA surveillance.



Involved in nonsense-mediated decay (NMD) of mRNAs



containing premature stop codons. Associates



preferentially with mRNAs produced by splicing. Does



not interact with pre-mRNAs, introns, or mRNAs



produced from intronless cDNAs. Associates with both



nuclear mRNAs and newly exported cytoplasmic



mRNAs. Complex with MAGOH is a component of the



nonsense mediated decay (NMD) pathway


Q9C0B6
Peptidylprolyl isomerase, FKBP-type
1.7
0.02


CHRFAM7A
After binding acetylcholine, the AChR responds by an
1.7
0.06



extensive change in conformation that affects all



subunits and leads to opening of an ion-conducting



channel across the plasma membrane


CYP3A4
Cytochromes P450 are a group of heme-thiolate
1.7
0.05



monooxygenases that perform a variety of oxidation



reactions.


EHD1
Acts in early endocytic membrane fusion and membrane
1.7
0.05



trafficking of recycling endosomes


ANKH
Regulates intra- and extracellular levels of inorganic
1.7
0.01



pyrophosphate (PPi), probably functioning as PPi



transporter


PAX4
Transcriptional repressor that binds to a common
1.7
0.02



element in the glucagon, insulin and somatostatin



promoters and plays an important role in the



differentiation and development of pancreatic islet beta



cells.


CACNA1B
Voltage-sensitive calcium channels (VSCC) mediate the
1.7
0.01



entry of calcium ions into excitable cells and are also



involved in a variety of calcium-dependent processes,



including muscle contraction, hormone or



neurotransmitter release, gene expression, cell motility,



cell division and cell death.


STX11
SNARE that acts to regulate protein transport between
1.7
0.03



late endosomes and the trans-Golgi network


NTN4
EGF-like domain; Laminin, N-terminal; Laminin-type
1.7
0.04



EGF-like domain; Netrin, C-terminal


Q9NUK6
Proteasome component region PCI
1.7
0.01


Q86UG6
Olfactory receptor; Rhodopsin-like GPCR superfamily
1.7
0.05


LILRB2
Receptor for class I MHC antigens. Recognizes a broad
1.7
0.00



spectrum of HLA-A, HLA-B, HLA-C and HLA-G



alleles. Involved in the down-regulation of the immune



response and the development of tolerance. Competes



with CD8A for binding to class I MHC antigens. Inhibits



FCGR1A-mediated phosphorylation of cellular proteins



and mobilization of intracellular calcium ions


NTRK2
Receptor for brain-derived neurotrophic factor (BDNF),
1.7
0.05



neurotrophin-3 and neurotrophin-4/5 but not nerve



growth factor (NGF). Involved in the development



and/or maintenance of the nervous system. This is a



tyrosine-protein kinase receptor. Known substrates for



the TRK receptors are SHC1, PI-3 kinase, and PLC-



gamma-1


Q8IYM2
Protein of unknown function DUF467
1.7
0.01


SART1
SART-1 protein
1.7
0.03


EDG4
Receptor for lysophosphatidic acid (LPA), a mediator of
1.7
0.03



diverse cellular activities. Seems coupled to the



G(i)/G(0), G(12)/G(13), and G(q) families of



heteromeric G proteins


O14562
Bipartite nuclear localization signal; Ubiquitin domain
1.7
0.01


Q8NC30
transmembrane
1.7
0.05


PLEK
Major protein kinase C substrate of platelets, its exact
1.7
0.05



function is not known


Q96C10
DEAD/DEAH box helicase; Helicase, C-terminal
1.7
0.01


SLC30A5
Cation efflux protein
1.7
0.02


RAX
Plays a critical role in eye formation by regulating the
1.7
0.01



initial specification of retinal cells and/or their



subsequent proliferation.


ABCE1
Antagonizes the binding of 2-5A (5′-phosphorylated
1.7
0.03



2′,5′;-linked oligoadenylates) by RNase L through direct



interaction with RNase L and therefore inhibits its



endoribonuclease activity. May play a central role in the



regulation of mRNA turnover. Antagonizes the anti-viral



effect of the interferon-regulated 2-5A/RNase L pathway


DHX9
Unwinds double-stranded DNA and RNA in a 3′ to 5′
1.7
0.05



direction. Alteration of secondary structure may



subsequently influence interactions with proteins or other



nucleic acids. Functions as a transcriptional activator


ZNF365
Zn-finger, C2H2 type
1.7
0.01


GGN
Proline-rich extensin; Proline-rich region
1.7
0.01


PRRX1
Acts as a transcriptional regulator of muscle creatine
1.7
0.04



kinase (MCK) and so has a role in the establishment of



diverse mesodermal muscle types. The protein binds to



an A/T-rich element in the muscle creatine enhancer


SLC7A7
Sodium-independent exchanger of cationic and large
1.7
0.01



neutral amino acids


Q9BYH8
Ankyrin
1.7
0.03


SMURF1
E3 ubiquitin-protein ligase which accepts ubiquitin from
1.7
0.03



an E2 ubiquitin-conjugating enzyme in the form of a



thioester and then directly transfers the ubiquitin to



targeted substrates. Interacts with receptor-regulated



SMADs specific for the BMP pathway, SMAD1 and



SMAD5, in order to trigger their ubiquitination and



degradation and hence their inactivation


ICAM3
ICAM proteins are ligands for the leukocyte adhesion
1.7
0.03



LFA-1 protein (integrin alpha-L/beta-2). ICAM3 is also



a ligand for integrin alpha-D/beta-2


C7orf23
Hypothetical protein C7orf23
1.7
0.05


SLC35B3
CGI-19 protein; chromosome 6 open reading frame 196.
1.7
0.02



solute carrier family 35, member B3


O00581
D111/G-patch domain; Forkhead-associated (FHA);
1.7
0.04



Proline-rich region; Ribosomal protein S5;


POLR2D
DNA-dependent RNA polymerase catalyzes the
1.7
0.02



transcription of DNA into RNA using the four



ribonucleoside triphosphates as substrates. Associates



with POLR2G


Q9NUQ9
NULL
1.7
0.05


STAU
Binds double-stranded RNA (regardless of the sequence)
1.7
0.05



and tubulin. May play a role in specific positioning of



mRNAs at given sites in the cell by crosslinking



cytoskeletal and RNA components, and in stimulating



their translation at the site


ZNF578
KRAB box; Zn-finger, C2H2 subtype;
1.7
0.03


FBXO21
Substrate-recognition component of the SCF (SKP1-
1.7
0.05



CUL1-F-box protein)-type E3 ubiquitin ligase complex


Q96LX7
Zn-finger, C2H2 type
1.7
0.06


MAPK12
Responds to activation by environmental stress and pro-
1.7
0.03



inflammatory cytokines by phosphorylating downstream



targets. Plays a role in myoblast differentiation and also



in the down-regulation of cyclin D1 in response to



hypoxia in adrenal cells suggesting MAPK12 may



inhibit cell proliferation while promoting differentiation


CLK3
Phosphorylates serine- and arginine-rich (SR) proteins of
1.7
0.05



the spliceosomal complex may be a constituent of a



network of regulatory mechanisms that enable SR



proteins to control RNA splicing. Phosphorylates serines,



threonines and tyrosines


RAP2C
ATP/GTP-binding site motif A (P-loop); Prenyl group
1.7
0.02



binding site (CAAX box); Ras GTPase superfamily


Q9ULQ0
low complexity
1.7
0.03


SPATA13
DH domain; Pleckstrin-like; Protamine P1; SH3 domain
1.7
0.01


GNA15
Guanine nucleotide-binding proteins (G proteins) are
1.7
0.05



involved as modulators or transducers in various



transmembrane signaling systems


IL12B
Cytokine that can act as a growth factor for activated T
1.7
0.00



and NK cells, enhance the lytic activity of



NK/lymphokine-activated killer cells, and stimulate the



production of IFN-gamma by resting PBMC


SSBP3
May be involved in transcription regulation of the alpha
1.7
0.05



2(I) collagen gene where it binds to the single-stranded



polypyrimidine sequences in the promoter region


ABCG4
May be involved in macrophage lipid homeostasis
1.7
0.01


SLAMF1
High-affinity self-ligand important in bidirectional T-cell
1.7
0.04



to B-cell stimulation. SLAM-induced signal-transduction



events in T lymphocytes are different from those in B



cells. Two modes of SLAM signaling are likely to exist:



one in which the inhibitor SH2D1A acts as a negative



regulator and another in which protein-tyrosine



phosphatase 2C-dependent signal transduction operates


C14orf166
Protein C14orf166
1.7
0.02


KPNA4
Functions in nuclear protein import as an adapter protein
1.7
0.03



for nuclear receptor KPNB1.


MXD3
Basic helix-loop-helix dimerization domain bHLH
1.6
0.04


CPSF6
Proline-rich extensin; Proline-rich region; RNA-binding
1.6
0.00



region RNP-1 (RNA recognition motif)


HAS1
Plays a role in hyaluronan/hyaluronic acid (HA)
1.6
0.00



synthesis. Also able to catalyze the synthesis of chito-



oligosaccharide depending on the substrate


SNX16
May be involved in several stages of intracellular
1.6
0.01



trafficking


URP2
Probably involved in cell adhesion
1.6
0.03


CAPZB
F-actin capping proteins bind in a Ca(2+)-independent
1.6
0.03



manner to the fast growing ends of actin filaments



(barbed end) thereby blocking the exchange of subunits



at these ends. Unlike other capping proteins (such as



gelsolin and severin), these proteins do not sever actin



filaments


Q8N4P3
Metal-dependent phosphohydrolase, HD region
1.6
0.05


MAP1A
Structural protein involved in the filamentous cross-
1.6
0.00



bridging between microtubules and other skeletal



elements


CHC1
Promotes the exchange of Ran-bound GDP by GTP.
1.6
0.03



Involved in the regulation of onset of chromosome



condensation in the S phase. Binds to the chromatin.



RCC1/Ran complex (together with other proteins) acts as



a component of a signal transmission pathway that



detects unreplicated DNA


PLXNA4
Cell surface receptor IPT/TIG; Plexin
1.6
0.05


CUL4A
Cullin
1.6
0.03


O94940
SAM (and some other nucleotide) binding motif
1.6
0.03


ABCA2
Probable transporter, unknown substrate. May have a
1.6
0.05



role in macrophage lipid metabolism and neural



development


C20orf135
Protein C20orf135
1.6
0.06


WASF1
Downstream effector molecules involved in the
1.6
0.05



transmission of signals from tyrosine kinase receptors



and small GTPases to the actin cytoskeleton


MAPK8IP3
The JNK-interacting protein (JIP) group of scaffold
1.6
0.02



proteins selectively mediates JNK signaling by



aggregating specific components of the MAPK cascade



to form a functional JNK signaling module. May



function as a regulator of vesicle transport, through



interations with the JNK-signaling components and



motor proteins


Q7M4L6
SH2 motif
1.6
0.01


EPS15
Involved in cell growth regulation. May be involved in
1.6
0.04



the regulation of mitogenic signals and control of cell



proliferation. Involved in the internalization of ligand-



inducible receptors of the receptor tyrosine kinase (RTK)



type, in particular EGFR


SFPQ
DNA- and RNA binding protein, involved in several
1.6
0.00



nuclear processes. Essential pre-mRNA splicing factor



required early in spliceosome formation and for splicing



catalytic step II


ECGF1
May have a role in maintaining the integrity of the blood
1.6
0.01



vessels. Has growth promoting activity on endothelial



cells, angiogenic activity in vivo and chemotactic



activity on endothelial cells in vitro


PCBD
Involved in tetrahydrobiopterin biosynthesis.
1.6
0.00


CASP4
Involved in the activation cascade of caspases
1.6
0.04



responsible for apoptosis execution. Cleaves caspase-1


HMGB1
Binds preferentially single-stranded DNA and unwinds
1.6
0.04



double stranded DNA


NUP62
Interleukin-4-induced protein 1 precursor; FIG.-1 protein
1.6
0.05


GLG1
Binds fibroblast growth factor and E-selectin (cell-
1.6
0.06



adhesion lectin on endothelial cells mediates binding of



neutrophils)


LILRA1
May act as receptor for class I MHC antigens; May act
1.6
0.02



as soluble receptor for class I MHC antigens


SUI1
Necessary for scanning and involved in initiation site
1.6
0.01



selection. Promotes the assembly of 48S ribosomal



complexes at the authentic initiation codon of a



conventional capped mRNA; Probably involved in



translation


TGDS
dTDP-D-glucose 4,6-dehydratase
1.6
0.01


Q9NPI0
low complexity; transmembrane
1.6
0.02


ZNF571
KRAB box; Zn-finger, C2H2 subtype
1.6
0.01


FGF10
Could be a growth factor active in the process of wound
1.6
0.04



healing. Acts as a mitogen in the lung. May act in a



manner similar to FGF-7


Q8N7I3
Immunoglobulin-like
1.6
0.01


GSG1
germ cell associated 1
1.6
0.06


KCNA10
K+ channel tetramerisation; Kv channel; Shaker voltage-
1.6
0.00



gated K+ channel


POLR1B
RNA polymerase beta subunit Rpb2, domain 2
1.6
0.05


IL32; NK4
May play a role in lymphocyte activation
1.6
0.00


O75121
Immunoglobulin-like
1.6
0.02


Q9H9C7
BRCT domain.
1.6
0.00


TNFRSF6
Receptor for TNFSF6/FASL. The adapter molecule
1.6
0.01



FADD recruits caspase-8 to the activated receptor. The



resulting death-inducing signaling complex (DISC)



performs caspase-8 proteolytic activation which initiates



the subsequent cascade of caspases (aspartate-specific



cysteine proteases) mediating apoptosis. FAS-mediated



apoptosis may have a role in the induction of peripheral



tolerance, in the antigen-stimulated suicide of mature T-



cells, or both. The secreted isoforms 2 to 6 block



apoptosis (in vitro)


EIF5A
The precise role of eIF-5A in protein biosynthesis is not
1.6
0.04



known but it functions by promoting the formation of the



first peptide bond


AREG
Bifunctional growth-modulating glycoprotein. Inhibits
1.6
0.02



growth of several human carcinoma cells in culture and



stimulates proliferation of human fibroblasts and certain



other tumor cells


DYRK1B
Dual-specificity kinase which possesses both serine/
1.6
0.05



threonine and tyrosine kinase activities. Enhances the



transcriptional activity of TCF1/HNF1A. Inhibits



epithelial cell migration.


Q96M86
Dynein heavy chain
1.6
0.01


KNS2
Kinesin is a microtubule-associated force-producing
1.6
0.00



protein that may play a role in organelle transport. The



light chain may function in coupling of cargo to the



heavy chain or in the modulation of its ATPase activity


SLC37A1
Glycerol-3-phosphate transporter; G-3-P transporter; G-
1.6
0.05



3-P permease; Solute carrier family 37 member 1


ICK
Protein kinase; Serine/Threonine protein kinase
1.6
0.02


Q8TEE6
ATP/GTP-binding site motif A (P-loop); Peptidase
1.6
0.00



family S16


Q8IUZ5
Aminotransferase class-III
1.6
0.06


HSPB1
Involved in stress resistance and actin organization
1.6
0.00


NFYA
Stimulates the transcription of various genes by
1.6
0.05



recognizing and binding to a CCAAT motif in



promoters, for example in type 1 collagen, albumin and



beta-actin genes


CLCA1
H+-transporting two-sector ATPase, gamma subunit;
1.6
0.03



von Willebrand factor, type A


SNTG1
Adapter protein that binds to and probably organizes the
1.6
0.02



subcellular localization of several proteins. May link



various receptors to the actin cytoskeleton and the



dystrophin glycol-protein complex. May participate in



regulating the subcellular location of diacylglycerol



kinase-zeta to ensure that diacyl-glycerol is rapidly



inactivated following receptor activation


TNFRSF1B
Receptor with high affinity for TNFSF2/TNF-alpha and
1.6
0.01



approximately 5-fold lower affinity for homotrimeric



TNFSF1/lymphotoxin-alpha. The TRAF1/TRAF2



complex recruits the apoptotic suppressors BIRC2 and



BIRC3 to TNFRSF1B/TNFR2. This receptor mediates



most of the metabolic effects of TNF-alpha. Isoform 2



blocks TNF-alpha-induced apoptosis, which suggests



that it regulates TNF-alpha function by antagonizing its



biological activity


C20orf85
Protein C20orf85
1.6
0.04


Q8N1Q9
Cation transporting ATPase, E1-E2 type
1.6
0.02


VGCNL1
Cation channel, non-ligand gated; Ion transport protein
1.6
0.03


FPGT
Catalyzes the formation of GDP-L-fucose from GTP and
1.6
0.00



L-fucose-1-phosphate. Functions as a salvage pathway to



reutilize L-fucose arising from the turnover of



glycoproteins and glycolipids


ENSG00000185305
ATP/GTP-binding site motif A (P-loop)
1.6
0.04


PDCD1LG1
Immunoglobulin-like
1.6
0.05


MFI2
Involved in iron cellular uptake. Seems to be internalized
1.6
0.01



and then recycled back to the cell membrane.


GK
Key enzyme in the regulation of glycerol uptake and
1.6
0.01



metabolism


Q9BVN4
Bacterial Sun/eukaryotic nucleolar Nop1/Nop2
1.6
0.06


NPB
May be involved in the regulation of neuroendocrine
1.6
0.03



system


VSX1
Binds to the 37-bp core of the locus control region
1.6
0.04



(LCR) of the red/green visual pigment gene cluster. May



regulate the activity of LCR


Q96N98
Amidase
1.6
0.04


Q96MB3
Protein kinase
1.6
0.04


PIM2
Serine/threonine-protein kinase Pim-2; Pim-2h
1.6
0.03


PHLDB1
Forkhead-associated (FHA); Pleckstrin-like
1.6
0.05


ENSG00000188719
unknown
1.6
0.06


ASB7
Ankyrin repeat and SOCS box protein 7; ASB-7
1.6
0.01


ENSG00000159752
low complexity; signal peptide
1.6
0.04


GABRA6
GABA, the major inhibitory neurotransmitter in the
1.6
0.04



vertebrate brain.


NMI
May be involved in augmenting coactivator protein
1.6
0.03



recruitment to a group of sequence-specific transcription



factors. Augments cytokine-mediated STAT



transcription. Enhances CBP/p300 coactivator protein



recruitment to STAT1 and STAT5


UNC5C
Death domain; Immunoglobulin-like; Thrombospondin,
1.6
0.02



type I; ZU5 domain


Q7Z2R6
low complexity; transmembrane
1.6
0.02


CNDP2; CN2;
Cytosolic nonspecific dipeptidase; Glutamate
1.6
0.04


CPGL
carboxypeptidase-like protein 1; CNDP dipeptidase 2


SF3B5
Pre-mRNA Splicing factor 3B subunit 5; 10 kDa subunit
1.6
0.04


DPYS
Dihydropyrimidinase; DHPase; Hydantoinase; DHP
1.6
0.01


DNAJA2
Co-chaperone of Hsc70
1.6
0.03


TPR
Component of the cytoplasmic fibrils of the nuclear pore
1.6
0.03



complex implicated in nuclear protein import. Its N-



terminus is involved in activation of oncogenic kinases


CD58
Ligand of the T lymphocyte CD2 glycoprotein. This
1.6
0.03



interaction is important in mediating thymocyte



interactions with thymic epithelial cells, antigen-



independent and -dependent interactions of T



lymphocytes with target cells and antigen-presenting



cells and the T lymphocyte rosetting with erythrocytes.



In addition, the LFA-3/CD2 interaction may prime



response by both the CD2+ and LFA-3+ cells


SLC22A11
General substrate transporter
1.5
0.05


Q9P1G3
Protein of unknown function DUF185
1.5
0.02


TIMM8B
Mitochondrial import inner membrane translocase
1.5
0.03



subunit TIM8 B; Deafness dystonia protein 2; DDP-like



protein


Q86W75
Sulfatase
1.5
0.01


Q9P2E5
chondroitin sulfate glucuronyltransferase
1.5
0.03


USP13
Ubiquitin carboxyl-terminal hydrolase 13; ISOT-3
1.5
0.05


NOTCH1
Functions as a receptor for membrane-bound ligands
1.5
0.04



Jagged1, Jagged2 and Delta1 to regulate cell-fate



determination. Upon ligand activation through the



released notch intracellular domain (NICD) it forms a



transcriptional activator complex with RBP-J kappa and



activates genes of the enhancer of split locus. Affects the



implementation of differentiation, proliferation and



apoptotic programs. May be important for normal



lymphocyte function. Involved in the maturation of both



CD4+ and CD8+ cells in the thymus


CHN2
GTPase-activating protein for p21-rac. Insufficient
1.5
0.03



expression of beta-2 chimaerin is expected to lead to



higher Rac activity and could therefore play a role in the



progression from low-grade to high-grade tumors


SS18
Synovial sarcoma, translocated to X chromosome
1.5
0.02



(SSXT, SYT)


TEP1
ATP/GTP-binding site motif A; G-protein beta WD-40
1.5
0.04



repeat


Q86XK7
Immunoglobulin-like; Myelin P0 protein
1.5
0.03


LPAL2
Chymotrypsin serine protease, family S1; Kringle;
1.5
0.04



Prothrombin; Serine protease, trypsin family


DHRS3
ATP/GTP-binding site motif A (P-loop); Glucose/ribitol
1.5
0.03



dehydrogenase; Insect alcohol dehydrogenase family;



Short-chain dehydrogenase/reductase SDR


PLAC1
Acc: NM_021796]; placenta-specific 1. [Source: RefSeq
1.5
0.01


Q96SV6;
ATP/GTP-binding site motif A (P-loop); Bipartite
1.5
0.05


Q9UJY0
nuclear localization signal; GTP-binding protein, HSR1-



related


RFX1
Regulatory factor essential for MHC class II genes
1.5
0.06



expression. Binds to the X boxes of MHC class II genes.



Also binds to an inverted repeat (ENH1) and to the most



upstream element (alpha) of the RPL30 promoter


SACS
May function in chaperone-mediated protein folding
1.5
0.01


FRMD4
Band 4.1 domain; Ezrin/radixin/moesin ERM
1.5
0.04


CCDC7
Coiled-coil domain containing 7.
1.5
0.05


SKP2
Substrate recognition component of the SCF (SKP1-
1.5
0.03



CUL1-F-box protein) E3 ubiquitin ligase complex which



mediates the ubiquitination and subsequent proteasomal



degradation of target proteins involved in cell cycle



progression, signal transduction and transcription.



Specifically recognizes phosphorylated



CDKN1B/p27kip and is involved in regulation of G1/S



transition.


PCDHGC3
Potential calcium-dependent cell-adhesion protein. May
1.5
0.06



be involved in the establishment and maintenance of



specific neuronal connections in the brain


WIF1
Binds to WNT proteins and inhibits their activities. May
1.5
0.01



be involved in mesoderm segmentation


RFC4
The elongation of primed DNA templates by DNA
1.5
0.04



polymerase delta and epsilon requires the action of the



accessory proteins proliferating cell nuclear antigen



(PCNA) and activator 1. The 37 kDa subunit may be



involved in the elongation of the multiprimed DNA



template


PFKFB3
Synthesis and degradation of fructose 2,6-bisphosphate
1.5
0.00


RGS1
Inhibits signal transduction by increasing the GTPase
1.5
0.03



activity of G protein alpha subunits thereby driving them



into their inactive GDP-bound form. This protein may be



involved in the regulation of B-cell activation and



proliferation


ZNF219
May function as a transcription factor
1.5
0.02


DAAM1
Binds to disheveled (Dvl) and Rho, and mediates Wnt-
1.5
0.00



induced Dvl-Rho complex formation. May play a role as



a scaffolding protein to recruit Rho-GDP and Rho-GEF,



thereby enhancing Rho-GTP formation


UBE2G2
Catalyzes the covalent attachment of ubiquitin to other
1.5
0.04



proteins


PPP1R15B
Protein phosphatase 1, regulatory subunit 15B.
1.5
0.02


PLEKHA3
Pleckstrin-like
1.5
0.06


GJA5
One gap junction consists of a cluster of closely packed
1.5
0.00



pairs of transmembrane channels, the connexons,



through which materials of low MW diffuse from cell to



neighboring cell


CACNA2D3
Cache domain; von Willebrand factor, type A
1.5
0.06


SDK1
Fibronectin, type III; Immunoglobulin-like
1.5
0.06


EIF2C2
Provides endonuclease activity to RNA-induced
1.5
0.04



silencing complexes (RISC). Cleaves siRNA/mRNA



heteroduplexes bound to RISC. Essential for embryonic



development as well as RNA-mediated gene silencing



(RNAi)


ARVCF
Involved in protein-protein interactions at adherens
1.5
0.06



junctions


HTLF
Binds to the purine-rich region in HTLV-I LTR
1.5
0.02


CD83
May play a significant role in antigen presentation or the
1.5
0.01



cellular interactions that follow lymphocyte activation


CST3
As an inhibitor of cysteine proteinases, this protein is
1.5
0.05



thought to serve an important physiological role as a



local regulator of this enzyme activity


PAPOLB
Polymerase that creates the 3′ poly(A) tail of mRNA's.
1.5
0.05



Also required for the endoribonucleolytic cleavage



reaction at some polyadenylylation sites. May acquire



specificity through interaction with a cleavage and



polyadenylation specificity factor (CPSF) at its C-



terminus


BIRC3
Apoptotic suppressor. The BIR motifs region interacts
1.5
0.02



with TNF receptor associated factors 1 and 2 (TRAF1



and TRAF2) to form an heteromeric complex, which is



then recruited to the tumor necrosis factor receptor 2



(TNFR2)


NPC1L1
Patched family
1.5
0.02


SERPINA1
Inhibitor of serine proteases. Its primary target is
1.5
0.05



elastase, but it also has a moderate affinity for plasmin



and thrombin


MATN4
Aspartic acid and asparagine hydroxylation site; EGF-
1.5
0.06



like calcium-binding; EGF-like domain; von Willebrand



factor, type A


Q9NWQ8
Phosphoprotein associated with glycosphingolipid-
1.5
0.03



enriched microdomains.


KCNJ2
Inward rectifier potassium channels are characterized by
1.5
0.01



a greater tendancy to allow potassium to flow into the



cell rather than out of it. Probably participates in



establishing action potential waveform and excitability



of neuronal/muscle tissues.


LATS2
Protein kinase; Serine/Threonine protein kinase;
1.5
0.05



Ubiquitin-associated domain


DHX16
Probable ATP-binding RNA helicase involved in pre-
1.5
0.05



mRNA splicing


TIGD4
CENP-B protein; CENP-B, N-terminal DNA-binding
1.5
0.06


CBX5
Component of heterochromatin. Recognizes and binds
1.5
0.05



histone H3 tails methylated at Lys-9, leading to



epigenetic repression. May interact with lamin B



receptor (LBR).


Q8TF25
Ankyrin; Sterile alpha motif SAM
1.5
0.02


KIAA0355
Hypothetical protein KIAA0355
1.5
0.00


SLC11A1
Divalent transition metal (iron and manganese)
1.5
0.02



transporter involved in iron metabolism and host



resistance to certain pathogens.


CUL5
Component of E3 ubiquitin ligase complexes, which
−1.5
0.04



mediate the ubiquitination and subsequent proteasomal



degradation of target proteins. May form a cell surface



vasopressin receptor


IL6R
Part of the receptor for interleukin 6. Binds to IL-6 with
−1.5
0.03



low affinity, but does not transduce a signal. Signal



activation necessitate an association with IL6ST.



Activation may lead to the regulation of the immune



response, acute-phase reactions and hematopoiesis


LY96
Cooperates with TLR4 in the innate immune response to
−1.5
0.03



bacterial lipopolysaccharide (LPS), and with TLR2 in the



response to cell wall components from Gram-positive



and Gram-negative bacteria. Enhances TLR4-dependent



activation of NFκB. Cells expressing both MD2 and



TLR4, but not TLR4 alone, respond to LPS


ENSG00000159797
low complexity
−1.5
0.03


ACVR1
On ligand binding, forms a receptor complex consisting
−1.5
0.02



of two type II and two type I transmembrane



serine/threonine kinases. Type II receptors phosphorylate



and activate type I receptors which autophosphorylate,



then bind and activate SMAD transcriptional regulators.



Receptor for activin


RNF8
Probable E3 ubiquitin-protein ligase required to
−1.5
0.05



ubiquitinate some nuclear proteins, and promote their



subsequent degradation


UACA
Ankyrin; Viral A-type inclusion protein repeat
−1.5
0.05


Q8WWN1
Protein kinase; SH3 domain; Serine/Threonine protein
−1.5
0.00



kinase; Tyrosine protein kinase


Q8TF72;
Bipartite nuclear localization signal; PDZ/DHR/GLGF
−1.5
0.04


Q96K23
domain; Proline-rich region


Q9BSW2
Calcium-binding EF-hand
−1.5
0.03


Q96L16
LOC200420
−1.5
0.01


TRIM26
Tripartite motif protein 26; Zinc finger protein 173; Acid
−1.5
0.00



finger protein; RING finger protein 95


WBP1
WW domain-binding protein 1; WBP-1
−1.5
0.02


CUL7
Component of a probable SCF-like E3 ubiquitin ligase
−1.5
0.00



complex, which mediates the ubiquitination and



subsequent proteosomal degaradation of target proteins.



Probably plays a role in the degradation of proteins



involved in endothelial proliferation and/or



differentiation


MCCC2
Methylcrotonoyl-CoA carboxylase beta chain,
−1.5
0.02



mitochondrial precursor


Q9Y5L9
HMG-I and HMG-Y DNA-binding domain (A + T-hook);
−1.5
0.05



Helicase, C-terminal; Proline-rich extensin; SNF2 related



domain


Q96GC0
Splicing factor 3b, subunit 3, 130 kD.
−1.5
0.02


FADD
Apoptotic adaptor molecule that recruits caspase-8 or
−1.5
0.05



caspase-10 to the activated Fas (CD95) or TNFR-1



receptors. The resulting aggregate called the death-



inducing signaling complex (DISC) performs caspase-8



proteolytic activation. Active caspase-8 initiates the



subsequent cascade of caspases (aspartate-specific



cysteine proteases) mediating apoptosis


MPP7
Guanylate kinase; L27 domain; PDZ/DHR/GLGF
−1.5
0.02



domain; SH3 domain


Q8N371
Transcription factor jumonji, jmjC
−1.5
0.06


UBE2R2
Ubiquitin-conjugating enzymes
−1.5
0.04


PGLYRP1
Binds specifically to peptidoglycan and is involved in
−1.5
0.04



innate immunity


Q96CR0
centrosome protein Cep63
−1.5
0.04


Q7Z3P6
G-protein beta WD-40 repeat; Proline-rich region
−1.5
0.02


Q9H8Y6
Bipartite nuclear localization signal
−1.5
0.03


C21orf33
ES1 protein homolog, mitochondrial precursor; KNP-I;
−1.5
0.01



GT335


SEC11L1
Part of signal peptidase complex, exact function
−1.5
0.04



unknown


Q8N5H3
Mouse Mammary Turmor Virus Receptor homolog 1.
−1.5
0.02


FER
Non-receptor tyrosine kinase. Probably performs
−1.5
0.00



important function, in regulatory processes such as cell



cycle control


Q8IUY5
Actin; Actin/actin-like
−1.5
0.02


Q96H61
low complexity
−1.5
0.02


C6orf208
unknown
−1.5
0.04


COL11A2
May play an important role in fibrillogenesis by
−1.5
0.05



controlling lateral growth of collagen II fibrils


PRRG1
Transmembrane proline-rich γ-carboxyglutamic acid
−1.5
0.04



protein 1


PPAP2A
PA-phosphatase related phosphoesterase
−1.5
0.01


TIMP2
Complexes with metalloproteinases (such as
−1.5
0.03



collagenases) and irreversibly inactivates them. Known



to act on MMP-1, MMP-2, MMP-3, MMP-7, MMP-8,



MMP-9, MMP-10, MMP-13, MMP-14, MMP-15,



MMP-16 and MMP-19


FHOD1
Required for the assembly of F-actin structures, such as
−1.5
0.01



stress fibers. Depends on the Rho-ROCK cascade for its



activity. Contributes to the coordination of microtubules



with actin fibers and plays a role in cell elongation


NOTCH1
Functions as a receptor for membrane-bound ligands
−1.5
0.05



Jagged1, Jagged2 and Delta1 to regulate cell-fate



determination. Upon ligand activation through the



released notch intracellular domain (NICD) it forms a



transcriptional activator complex with RBP-Jκ and



activates genes of the enhancer of split locus. Affects the



implementation of differentiation, proliferation and



apoptotic programs. May be important for normal



lymphocyte function. In altered form, may contribute to



transformation or progression in some T-cell neoplasms.



Involved in the maturation of both CD4+ and CD8+ cells



in the thymus.


HINT3
Histidine triad (HIT) protein
−1.5
0.04


POLR2L
DNA-dependent RNA polymerase
−1.5
0.03


ZNF354A
Transcription factor zinc finger protein 354A, 17, eZNF
−1.5
0.05


INPP5D
Endonuclease/exonuclease/phosphatase family; Proline-
−1.5
0.00



rich extensin; SH2 motif


C9orf37
low complexity
−1.5
0.02


HPGD
Inactivation of prostaglandins
−1.5
0.05


Q8WYL1
Dual specificity protein phosphatase
−1.5
0.01


GBX1
Homeobox protein GBX-1; Gastrulation and brain-
−1.5
0.05



specific homeobox protein 1


ZCCHC4
ATP/GTP-binding site motif A (P-loop); Bipartite
−1.6
0.04



nuclear localization signal; N-6 Adenine-specific DNA



methylase; Zn-finger, CCHC type; Zn-finger, DHHC



type


Q8N3J9
Zn-finger, C2H2 subtype;
−1.6
0.00


CBX1
Component of heterochromatin. Recognizes and binds
−1.6
0.04



histone H3 tails methylated at Lys-9, leading to



epigenetic repression. May interact with lamin B



receptor (LBR).


PARD3
Adapter protein involved in asymmetrical cell division
−1.6
0.05



and cell polarization processes. Seems to play a central



role in the formation of epithelial tight junctions.


SERPINA10
Inhibits factor Xa activity in the presence of protein Z,
−1.6
0.05



calcium and phospholipid


CENTG3
GTPase-activating protein for the ADP ribosylation
−1.6
0.06



factor family


ARHGEF19
Bipartite nuclear localization signal; DH domain;
−1.6
0.05



Pleckstrin-like; SH3 domain


Q8IWC2
ATP/GTP-binding site motif A (P-loop); Proline-rich
−1.6
0.02



extensin


TCP10
T-complex protein 10A homolog
−1.6
0.04


ENSG00000170714
low complexity
−1.6
0.04


TPCN1
Ion transport protein; Legume lectin, beta domain
−1.6
0.03


APP
Functions as a cell surface receptor. Involved in cell
−1.6
0.05



mobility and transcription regulation through protein-



protein interactions. Can promote transcription activation



through binding to APBB1/Tip60 and inhibit Notch



signaling through interaction with Numb. Couples to



apoptosis-inducing pathways such as those mediated by



G(O) and JIP.


IDI1
Catalyzes the 1,3-allylic rearrangement of the
−1.6
0.00



homoallylic substrate isopentenyl (IPP) to its highly



electrophilic allylic isomer, dimethylallyl diphosphate



(DMAPP)


EZH1
May be involved in the regulation of gene transcription
−1.6
0.06



and chromatin structure


ASB7
Ankyrin repeat and SOCS box protein 7; ASB-7
−1.6
0.04


ACTL7A
Actin-like protein 7A; Actin-like-7-alpha; Actin-like 7A
−1.6
0.00


GAPD
Glyceraldehyde-3-phosphate dehydrogenase; GAPDH
−1.6
0.01


SULT1E1
May control the level of the estrogen receptor by
−1.6
0.01



sulfurylating free estradiols etc.


Q9BV79
Zinc-containing alcohol dehydrogenase superfamily
−1.6
0.05


C6orf110
Protein of unknown function DUF221
−1.6
0.03


Q8IXK7
low complexity; transmembrane
−1.6
0.02


NRCAM
Cell adhesion, ankyrin-binding protein involved in
−1.6
0.05



neuron-neuron adhesion.


ACTA2
Actins are highly conserved proteins that are involved in
−1.6
0.04



various types of cell motility and are ubiquitously



expressed in all eukaryotic cells


Q9BPX8
unknown
−1.6
0.02


Q9HCK1
low complexity
−1.6
0.00


STK29
BR serine/threonine-protein kinase 2, 29; SAD1B
−1.6
0.01


MAGEA9
May play a role in embryonal development and tumor
−1.6
0.05



transformation or aspects of tumor progression


GCAT
2-amino-3-ketobutyrate coenzyme A ligase,
−1.6
0.01



mitochondrial


NPAS2
Neuronal PAS domain protein 2; Member of PAS
−1.6
0.00



protein 4; MOP4


GLRX
Has a glutathione-disulfide oxidoreductase activity in the
−1.6
0.04



presence of NADPH and glutathione reductase.


CLECSF6
C-type lectin; Type II antifreeze protein
−1.6
0.02


OSBPL11
Oxysterol binding protein-related protein 11
−1.6
0.00


ANKRD10
Ankyrin repeat domain protein 10
−1.6
0.04


SH3MD3
SH3 domain
−1.6
0.01


SFRP5
Bipartite nuclear localization signal; Frizzled CRD
−1.6
0.05



region; Netrin, C-terminal


Q8IZW8
SH2 motif
−1.6
0.02


OVOL1
Putative transcription factor. Involved in hair formation
−1.6
0.02



and spermatogenesis. May function in the differentiation



and/or maintenance of the urogenital system


INSR
This receptor binds insulin and has a tyrosine-protein
−1.6
0.01



kinase activity. Isoform Short has a higher affinity for



insulin


RBPSUHL
Putative transcription factor, which cooperates with
−1.6
0.03



EBNA2 to activate transcription


PRM2
Protainines substitute for histones in the chromatin of
−1.6
0.02



sperm during the haploid phase of spermatogenesis &



compact sperm DNA into a highly condensed, stable and



inactive complex


PARVA
Probably plays a role in the regulation of cell adhesion
−1.6
0.03



and cytoskeleton organization


VPS4B
Involved in intracellular protein transport probably out of
−1.6
0.04



a prevacuolar endosomal compartment. May be involved



in the release of components of the bilayered coat from



the endosomal membrane. In case of infection, the HIV-



1 virus takes advantage of it for budding and exocytic



cargoes of viral proteins


APRIN
HMG-I and HMG-Y DNA-binding domain (A + T-hook)
−1.6
0.04


Q96MI8
ATP/GTP-binding site motif A (P-loop)
−1.6
0.06


CDH8
Cadherins are calcium dependent cell adhesion proteins.
−1.6
0.04



They preferentially interact with themselves in a



homophilic manner in connecting cells; cadherins may



thus contribute to the sorting of heterogeneous cell types


PIK3C2B
Phosphorylates PtdIns and PtdIns4P with a preference
−1.6
0.05



for PtdIns. Does not phosphorylate PtdIns(4,5)P2. May



be involved in EGF and PDGF signaling cascades


CALD1
Actin- and myosin-binding protein implicated in the
−1.6
0.05



regulation of actomyosin interactions in smooth muscle



and nonmuscle cells (could act as a bridge between



myosin and actin filaments). Also plays an essential role



during cellular mitosis and receptor capping.


ARRDC1
Arrestin
−1.6
0.01


Q9BWC9
Bipartite nuclear localization signal
−1.6
0.01


MC4R
Receptor specific to the heptapeptide core common to
−1.6
0.02



adrenocorticotropic hormone and alpha-, beta-, and



gamma-MSH. This receptor is mediated by G proteins



that stimulate adenylate cyclase


ELMO1
Involved in cytoskeletal rearrangements required for
−1.6
0.03



phagocytosis of apoptotic cells and cell motility. Acts in



assocation with DOCK1 and CRK. Was initially



proposed to be required in complex with DOCK1 to



activate Rac Rho small GTPases. May enhance the



guanine nucleotide exchange factor (GEF) activity of



DOCK1


MPDZ
PDZ/DHR/GLGF domain
−1.6
0.06


ENSG00000079548
low complexity
−1.6
0.06


FECH
Catalyzes the ferrous insertion into protoporphyrin IX
−1.6
0.01


PNMA2
Paraneoplastic antigen Ma2
−1.6
0.04


KCNMB4
Calcium-activated BK potassium channel, beta subunit
−1.6
0.04


STAG1
Component of cohesin complex, a complex required for
−1.6
0.05



the cohesion of sister chromatids after DNA replication.


MPPE1
Hemopexin repeat; Metallo-phosphoesterase
−1.6
0.02


Q8N6S2
Immunoglobulin-like; N-6 Adenine-specific DNA
−1.6
0.06



methylase


UBR1
Polyprenyl synthetase; Ribosomal protein S16; Zn-finger
−1.6
0.01



(putative), N-recognin


WASF2
Actin-binding WH2; Bipartite nuclear localization
−1.6
0.02



signal; Histamine H3 receptor; Pistil-specific extensin-



like protein; Proline-rich extensin


ABR
GTPase-activating protein for RAC and CDC42.
−1.6
0.01



Promotes the exchange of RAC or CDC42-bound GDP



by GTP, thereby activating them


CYP1B1
Cytochromes P450 are a group of heme-thiolate
−1.6
0.02



monooxygenases. In liver microsomes, this enzyme is



involved in an NADPH-dependent electron transport



pathway. It oxidizes a variety of structurally unrelated



compounds, including steroids, fatty acids, and



xenobiotics


EEF2K
Phosphorylates eukaryotic elongation factor-2. Binds
−1.6
0.03



calmodulin


HIST1H2BN
Histone H2B
−1.6
0.00


BRD3
Bromodomain-containing protein 3; RING3-like protein
−1.6
0.04


MFN1
ATP/GTP-binding site motif A (P-loop); Fzo-like
−1.6
0.05



conserved region


C11orf21
Protein C11orf21
−1.6
0.00


ZFP64
May function as a transcription factor
−1.6
0.02


PLA2G5
PA2 catalyzes the calcium-dependent hydrolysis of the
−1.6
0.02



2-acyl groups in 3-sn-phosphoglycerides. May be



involved in the production of lung surfactant, the



remodeling or regulation of cardiac muscle


Q96HP4
Flavoprotein pyridine nucleotide cytochrome reductase;
−1.6
0.00



NADH: cytochrome b5 reductase (CBR); Oxidoreductase



FAD/NAD(P)-binding; Phenol hydroxylase reductase


MYOZ2
FATZ related protein 2; calcineurin-binding protein
−1.6
0.04



calsarcin-1


GOLGB1
May participate in forming intercisternal cross-bridges of
−1.6
0.04



the Golgi complex


Q96HQ2
low complexity
−1.6
0.06


TESK2
Dual specificity protein kinase activity catalyzing
−1.6
0.02



autophosphorylation and phosphorylation of exogenous



substrates on both serine/threonine and tyrosine residues.



Phosphorylates cofilin at Ser-3.


AK2
This small ubiquitous enzyme is essential for
−1.6
0.01



maintenance and cell growth


Q9C0D3
Bipartite nuclear localization signal; Leucine-rich repeat
−1.6
0.03


ZNF347
Bipartite nuclear localization signal; KRAB box; Zn-
−1.6
0.05



finger, C2H2 subtype


MUC20
ATP/GTP-binding site motif A (P-loop)
−1.7
0.04


CLECSF14
C-type lectin; Type II antifreeze protein
−1.7
0.01


ECM1
Extracellular matrix protein 1 precursor; Secretory
−1.7
0.03



component p85


Q9P021
HSPC139 protein; postsynaptic protein CRIPT
−1.7
0.01


Q9P194
unknown
−1.7
0.04


Q96CD2
Flavoprotein
−1.7
0.02


STMN4
Stathmin-4; Stathmin-like protein B3; RB3
−1.7
0.06


APTX
Histidine triad (HIT) protein; Zn-finger, C2H2 type.
−1.7
0.05


Q8IYX7
unknown
−1.7
0.01


CXXC5
Bipartite nuclear localization signal; Zn-finger, CXXC
−1.7
0.02



type


PPM1A
Enzyme with a broad specificity
−1.7
0.02


Q9P2I9
Bipartite nuclear localization signal
−1.7
0.04


Q9BSD4
coiled-coil; low complexity
−1.7
0.05


THAP9
Bipartite nuclear localization signal
−1.7
0.05


PLXDC2
Plexin
−1.7
0.01


Q9NTK9
DJ1092A11.1 (Hypothetical protein KIAA0495).
−1.7
0.04


MTIF2
One of the essential components for the initiation of
−1.7
0.00



protein synthesis.


RNF122
Zn-finger, RING
−1.7
0.04


CRYZL1
Quinone oxidoreductase-like 1; Zeta-crystallin homolog;
−1.7
0.04



4P11


SLC39A10
Cytochrome c heme-binding site; Zinc transporter ZIP
−1.7
0.03


AKAP11
Binds to the N-terminal PTS2-type peroxisomal targeting
−1.7
0.02



signal and plays an essential role in peroxisomal protein



import; Binds to type II regulatory subunits of protein



kinase A and anchors/targets them to the membrane.



May anchor the kinase to cytoskeletal and/or organelle-



associated proteins; Ligand of the T lymphocyte CD2



glycoprotein.


TNNC2
Troponin is the central regulatory protein of striated
−1.7
0.02



muscle contraction. Tn consists of three components: Tn-



I which is the inhibitor of actomyosin ATPase, Tn-T



which contains the binding site for tropomyosin and Tn-



C. The binding of calcium to Tn-C abolishes the



inhibitory action of Tn on actin filaments


TTC17
TPR repeat
−1.7
0.01


NPTX1
May mediate uptake of degraded synaptic material which
−1.7
0.01



could play an important role in synaptic remodeling.


FMNL2
Actin-binding FH2; Proline-rich extensin; Wilm's
−1.7
0.01



tumour protein


HLA-DMA
Plays a critical role in catalyzing the release of class II
−1.7
0.01



HLA-associated invariant chain-derived peptides (CLIP)



from newly synthesized class II HLA molecules and



freeing the peptide binding site for acquisition of



antigenic peptides


SLC22A13
General substrate transporter
−1.7
0.05


H2AFZ
Variant histones H2A are synthesized throughout the cell
−1.7
0.00



cycle and are very different from classical S-phase



regulated H2A. The exact function of variant histones



H2A is not known


HNRPM
Pre-mRNA binding protein in vivo, binds avidly to
−1.7
0.01



poly(G) and poly(U) RNA homopolymers in vitro.



Involved in splicing. Acts as a receptor for



carcinoembryonic antigen in Kupffer cells, may initiate a



series of signaling events leading to tyrosine



phosphorylation of proteins and induction of IL-1 alpha,



IL-6, IL-10 and tumor necrosis factor alpha cytokines


OR5A2
Putative odorant receptor
−1.7
0.04


SV2B
General substrate transporter; Sugar transporter
−1.7
0.00



superfamily


DNTTIP1
Shown to enhance TdT activity, in vitro
−1.7
0.04


HSPA1L
In cooperation with other chaperones, Hsp70s stabilize
−1.7
0.02



preexistent proteins against aggregation and mediate the



folding of newly translated polypeptides in the cytosol as



well as within organelles.


KIAA0258
Protein KIAA0258
−1.7
0.02


MRPS16
28S ribosomal protein S16, mitochondrial precursor
−1.7
0.04


ITGA10
Integrin alpha-10/beta-1 is a receptor for collagen
−1.7
0.05


IL10RB
Receptor for IL10 and IL22. Serves as an accessory
−1.7
0.02



chain essential for the active IL10 receptor complex and



to initiate IL10-induced signal transduction events


Q9C0D5
ATP/GTP-binding site motif A (P-loop); Ankyrin; TPR
−1.7
0.04



repeat


C21orf57
UPF0054 protein C21orf57
−1.7
0.03


TMH
unknown
−1.7
0.05


PLA2G4C
Lysophospholipase, catalytic domain; Prenyl group
−1.7
0.01



binding site (CAAX box)


PPAT
Glutamine phosphoribosylpyrophosphate
−1.7
0.01



amidotransferase


Q7Z570
Zn-finger, C2H2 matrin type
−1.7
0.06


ARL3
Does not act as an allosteric activator of the cholera toxin
−1.7
0.02



catalytic subunit


PHF5A
Acts as a transcriptional regulator by binding to the
−1.7
0.04



GJA1/Cx43 promoter and enhancing its up-regulation by



ESR1/ER-alpha. Also involved in pre-mRNA splicing


MTHFR
Catalyzes the conversion of 5,10-
−1.7
0.02



methylenetetrahydrofolate to 5-methyltetrahydrofolate, a



co-substrate for homocysteine remethylation to



methionine


MARK4
MAP/microtubule affinity-regulating kinase 4
−1.8
0.05


MVP
Unknown, though MVP is required for normal vault
−1.8
0.05



structure. Vaults are multi-subunit structures that may be



involved in nucleo-cytoplasmic transport


USP6NL
RabGAP/TBC domain
−1.8
0.02


PAPPA
Metalloproteinase which specifically cleaves IGFBP-4
−1.8
0.04



and IGFBP-5, releasing bound IGF. Cleavage of IGFBP-



4 is dramatically enhanced by IGF, whereas cleavage of



IGFBP-5 is slightly inhibited by the presence of IGF


RAB27B
Ras-related protein Rab-27B; C25KG
−1.8
0.02


BNIP3L
Induces apoptosis. Interacts with viral and cellular anti-
−1.8
0.05



apoptosis proteins. Can overcome the suppressers BCL-2



and BCL-XL, although high levels of BCL-XL



expression will inhibit apoptosis. May function as a



tumor suppressor


ENSG00000187712
Actin/actin-like
−1.8
0.03


C6orf80
low complexity
−1.8
0.02


Q96EG4
low complexity
−1.8
0.06


095893
transmembrane
−1.8
0.04


COQ4
Ubiquinone biosynthesis protein COQ4 homolog;
−1.8
0.01


DDC
Catalyzes the decarboxylation of DOPA to dopamine, L-
−1.8
0.04



5-hydroxytryptophan to serotonin and L-trp to



tryptamine


ZNF431
May function as a transcription factor
−1.8
0.03


JAK2
Tyrosine kinase of the non-receptor type, involved in
−1.8
0.03



interleukin 3 signal transduction


RNF34
Cytochrome c heme-binding site; Zn-finger, RING
−1.8
0.02


Q9BYA4
unknown
−1.8
0.06


ATP1B4
This is the non-catalytic component of a yet unknown
−1.8
0.03



soduium or proton exchange ATPase


PCDHGC3
Potential calcium-dependent cell-adhesion protein.
−1.8
0.03


C6orf33
Hly-III related proteins
−1.8
0.02


Q96BY9
NULL
−1.8
0.04


Q9BZS9
PNAS-138.
−1.8
0.03


Q96K66
signal peptide
−1.8
0.02


Q9Y4M2
low complexity
−1.9
0.02


CDC14B
Tyrosine specific protein phosphatase; Tyrosine specific
−1.9
0.04



protein phosphatase and dual specificity protein



phosphatase


CALM3
Calmodulin mediates the control of a large number of
−1.9
0.05



enzymes by Ca(2+). Among the enzymes to be



stimulated by the calmodulin-Ca(2+) complex are a



number of protein kinases and phosphatases


SLC16A9
Solute carrier family 16 (monocarboxylic acid
−1.9
0.05



transporters), member 9


Q8TEB9
Rhomboid-like protein; Ubiquitin interacting motif
−1.9
0.02


Q9UPP5
coiled-coil; low complexity
−1.9
0.01


Q8IUT6
low complexity; transmembrane
−1.9
0.01


ARRB1
Beta-arrestins seem to bind phosphorylated beta-
−1.9
0.03



adrenergic receptors and regulate function, thereby



causing a significant impairment of their capacity to



activate G(S) proteins


ATP6V1H
Subunit of the peripheral V1 complex of vacuolar
−1.9
0.00



ATPase. Subunit H activate ATPase activity of the



enzyme and couple ATPase activity to proton flow.



Vacuolar ATPase is responsible for acidifying a variety



of intracellular compartments in eukaryotic cells.



Involved in the endocytosis mediated by clathrin-coated



pits, required for the formation of endosomes


Q969E4
Bipartite nuclear localization signal
−1.9
0.02


GBAS
Protein NipSnap2; Glioblastoma amplified sequence
−1.9
0.03


MYCL1
L-myc-1 proto-oncogene protein
−1.9
0.04


C22orf3
Protein C22orf3
−1.9
0.00


EPHX2
Acts on epoxides (alkene oxides, oxiranes) and arene
−1.9
0.03



oxides. Also determines steady-state levels of



physiological mediators.


ZFP36L2
Probable regulatory protein involved in regulating the
−1.9
0.04



response to growth factors


Q9NS00
Acc: NM_020156]; core1 UDP-galactose: N-
−1.9
0.03



acetylgalactosamine-alpha-R beta 1,3-galac.



[Source:RefSeq


O95510
ABC transporter; Acyl transferase domain
−1.9
0.00


Q8TB55
Proline-rich region
−1.9
0.00


Q9Y3S6
RhoGAP domain
−1.9
0.03


FER
Tyrosine kinase of the non-receptor type. Probably
−1.9
0.05



performs an important function, perhaps in regulatory



processes such as cell cycle control


Q8NAA4
G-protein beta WD-40 repeat
−1.9
0.00


ENSG00000115404
Enoyl-CoA hydratase/isomerase
−2.0
0.04


DHRS9
Glucose/ribitol dehydrogenase; Short-chain
−2.0
0.05



dehydrogenase/reductase SDR


ELOVL5
GNS1/SUR4 membrane protein
−2.0
0.01


IL13RA1
Binds IL13 with a low affinity. Together with IL4R-
−2.0
0.00



alpha can form a functional receptor for IL13. Also



serves as an alternate accessory protein to the common



cytokine receptor gamma chain for IL4 signaling, but



cannot replace the function of gamma C in allowing



enhanced IL2 binding activity


Q8IXM2
low complexity
−2.0
0.05


CYB5
Cytochrome b5 is a membrane bound hemoprotein that
−2.0
0.01



functions as an electron carrier for several membrane



bound oxygenases


MAP1B
Phosphorylated MAP1B may play a role in the
−2.0
0.04



cytoskeletal changes that accompany neurite extension.



Possibly MAP1B Binds to at least two tubulin subunits



in the polymer, and this bridging of subunits might be



involved in nucleating microtubule polymerization and



in stabilizing microtubules


CABP2
Calcium-binding protein 2; CaBP2
−2.0
0.01


Q8TDG4
DEAD/DEAH box helicase; Helicase, C-terminal
−2.0
0.05


FAM31B
DENN (AEX-3) domain; Proline-rich extensin; dDENN
−2.0
0.04



domain


KIAA1244
Essential component of the high affinity receptor for the
−2.0
0.04



general membrane fusion machinery and an important



regulator of transport vesicle docking and fusion


CDK2AP1
Cyclin-dependent kinase 2-associated protein 1; Putative
−2.0
0.06



oral cancer suppressor.


C5orf5
Protein C5orf5; GAP-like protein N61
−2.0
0.03


MOCS3
Activates MPT synthase by the ATP dependant
−2.1
0.05



adenylation of its C-terminal residue


UGT2B7
Heavy metal transport/detoxification protein; UDP-
−2.1
0.01



glucoronosyl/UDP-glucosyl transferase


ARL7
Binds and exchanges GTP and GDP
−2.1
0.00


CD4
Accessory protein for MHC class-II antigen/T-cell
−2.1
0.01



receptor interaction. May regulate T-cell activation


KRT7
Keratin, type II cytoskeletal 7; Cytokeratin-7;
−2.1
0.05



Sarcolectin


ENSG00000142954
4Fe—4S ferredoxin, iron-sulfur binding domain;
−2.1
0.01



Dihydroorotate dehydrogenase; FMN/related compound-



binding core


Q96FP9
Calponin-like actin-binding; Leucine-rich repeat
−2.1
0.05


GNAS
The G(s) Guanine nucleotide-binding protein is involved
−2.1
0.04



in hormonal regulation of adenylate cyclase: it activates



the cyclase in response to beta-adrenergic stimuli


Q9P1V9
low complexity
−2.1
0.01


Q9Y547
HSPCO34 protein.
−2.2
0.02


ENSG00000177876
Thrombospondin, type I
−2.2
0.02


Q9BR68
Ran-interacting Mog1 protein
−2.2
0.03


CLGN
Probably plays an important role in spermatogenesis.
−2.2
0.01



Binds calcium ions


Q96FV0
Leucine-rich repeat
−2.2
0.01


PLAGL2
Shows weak transcriptional activatory activity
−2.2
0.04


ITM2B
Integral membrane protein 2B; Transmembrane protein
−2.2
0.06



BRI; ABri/ADan amyloid peptide


Q8IY68
low complexity
−2.2
0.03


SLC1A4
Transporter for alanine, serine, cysteine, and threonine.
−2.3
0.05



Exhibits sodium dependence


TIGD7
CENP-B protein; CENP-B, N-terminal DNA-binding
−2.3
0.02


VTI1A
V-SNARE that mediates vesicle transport pathways
−2.3
0.06



through interactions with t-SNAREs on the target



membrane. These interactions are proposed to mediate



aspects of the specificity of vesicle trafficking and to



promote fusion of the lipid bilayers. May be concerned



with increased secretion of cytokines associated with



cellular senescence


ITGBL1
Integrin beta, C-terminal; Laminin-type EGF-like
−2.3
0.01



domain


Q9C093
ATP/GTP-binding site motif A (P-loop)
−2.3
0.03


FOS
Nuclear phosphoprotein which forms a tight but non-
−2.3
0.00



covalently linked complex with the JUN/AP-1



transcription factor. In the heterodimer, c-fos and



JUN/AP-1 basic regions each seems to interact with



symmetrical DNA half sites. C-fos has a critical function



in regulating the development of cells destined to form



and maintain the skeleton. it is thought to have an



important role in signal transduction, cell proliferation



and differentiation


LARS
Aminoacyl-tRNA synthetase, class Ia
−2.5
0.01


Q96FR9
Bipartite nuclear localization signal; Exonuclease
−2.5
0.01


P2RY5
P2Y purinoceptor 5; P2Y5;
−2.5
0.01


SLC26A10
DH domain
−2.5
0.02


PHTF1
May play a role in transcription regulation
−2.6
0.00


DEFB127
Has antibacterial activity
−2.8
0.00


MEF2C
Transcription activator which binds specifically to the
−2.8
0.04



MEF2 element present in the regulatory regions of many



muscle-specific genes


DBR1
Lariat debranching enzyme, C-terminal; Metallo-
−2.8
0.05



phosphoesterase


DHRS8
2,3-dihydro-2,3-dihydroxybenzoate dehydrogenase;
−3.0
0.02



Short-chain dehydrogenase/reductase SDR


O75915
Prenylated rab acceptor PRA1
−3.3
0.03


CD36
Seems to have numerous potential physiological
−5.8
0.05



functions. Binds to collagen, thrombospondin, anionic



phospholipids and oxidized LDL. May function as a cell



adhesion molecule. Directly mediates cytoadherence of



Plasmodium falciparum parasitized erythrocytes. Binds



long chain fatty acids and may function in the transport



and/or as a regulator of fatty acid transport


RBP1
Intracellular transport of retinol
125.8
0.05
















TABLE 72










Thirty nine genes that were up-regulated by LPS and somewhat suppressed in


the presence of peptide SEQ ID NO: 7. Such genes reflect the anti-endotoxic activity of


SEQ ID NO: 7. The data reveal that the residual levels of expression can be largely


accounted for by the response to peptide SEQ ID NO: 7 alone.




















Fold





Fold
p-
Fold
p-
Change


Gene

change
student
change
student
by
p-student


Name
Gene Description
by LPS
(LPS)
by LPS + peptide
(LPS + peptide)
peptide
(peptide)

















GPD1
Cytoplasmic glycerol-3-phosphate
103.4
0.06
89.7
0.04
65.3
0.09



dehydrogenase [NAD+],


Q8NI35
ATP/GTP-binding site motif A (P-
78.4
0.06
53.9
0.06
57.2
0.05



loop);


FEZ2
Involved in axonal outgrowth and
45.9
0.05
25.8
0.02
40.2
0.02



fasciculation


NRXN1
Neuronal cell surface protein that may
35.4
0.05
25.9
0.04
31.9
0.09



be involved in cell recognition and cell



adhesion. May mediate intracellular



signaling


PLCG1
PLC-gamma is a major substrate for
32.6
0.04
17.0
0.04
18.4
0.05



heparin-binding growth factor 1 (acidic



fibroblast growth factor)-activated



tyrosine kinase


Q7RTU0
Basic helix-loop-helix dimerization
21.4
0.03
8.4
0.06
10.4
0.05



domain bHLH


ALDOB
Fructose-bisphosphate aldolase B;
21.0
0.03
15.5
0.01
8.9
0.06



Liver-type aldolase


Q9H5P1
Zn-finger, C-x8-C-x5-C-x3-H type
19.9
0.06
8.6
0.02
9.1
0.05


SYT11
May be involved in Ca(2+)-dependent
14.8
0.03
6.1
0.06
7.3
0.08



exocytosis of secretory vesicles


UBXD2
UBX domain-containing protein 2
9.3
0.04
6.8
0.04
5.6
0.04


PROZ
Appears to assist hemostasis by
6.9
0.05
2.4
0.05
4.0
0.27



binding thrombin and promoting its



association with phospholipid vesicles


PLAC8
Placenta-specific gene 8 protein; C15
6.4
0.01
2.2
0.05
1.6
0.04



protein


Q96PN6
ATP/GTP-binding site motif A (P-
5.2
0.05
3.3
0.03
3.0
0.08



loop); Guanylate cyclase


ASTN2
Fibronectin, type III
5.1
0.03
3.7
0.06
4.0
0.16


O60290
KRAB box
4.8
0.05
2.9
0.04
4.2
0.08


FTCD
Folate-dependent enzyme, that has
4.6
0.03
3.8
0.03
4.9
0.06



transferase and deaminase activity.



Serves to channel one-carbon units to



the folate pool


NFKB2
NFκB subunits p52 and p100
4.3
0.02
2.8
0.01
1.9
0.15


CTLA4
Possibly involved in T-cell activation.
4.1
0.02
2.7
0.01
2.4
0.13



Binds to B7-1 (CD80) and B7-2



(CD86)


PSMA1
Proteasome subunit
4.1
0.04
2.7
0.04
3.8
0.04


CCL2
Chemotactic factor that attracts
3.4
0.02
2.9
0.01
1.3
0.10



monocytes and basophils but not



neutrophils or eosinophils.


HNF4A
Transcriptionally controlled
3.3
0.01
2.6
0.01
3.0
0.07



transcription factor. Binds to DNA



sites required e.g. for the transcription



of alpha 1-antitrypsin, and HNF1-



alpha.


MAFF
Interacts with the upstream promoter
3.3
0.01
2.5
0.00
2.1
0.04



region of the oxytocin receptor gene.



May be involved in the cellular stress



response


FBXO32
Probably recognizes and binds to some
3.3
0.03
1.9
0.02
1.2
0.11



phosphorylated proteins and promotes



their ubiquitination and degradation



during skeletal muscle atrophy


TNF
Cytokine tumour necrosis factor α
3.3
0.02
2.4
0.01
1.2
0.82


NPAS2
Neuronal PAS domain protein 2;
2.8
0.00
1.8
0.03
2.4
0.04


ICAM3
Ligands for the leukocyte adhesion
2.6
0.04
1.7
0.03
2.2
0.05



LFA-1 protein and integrin alpha-



D/beta-2


Q8NC30
transmembrane
2.3
0.00
1.7
0.05
1.2
0.82


Q8IUC6
Proline-rich extensin domain
2.3
0.03
1.4
0.05
1.3
0.28


O94940
SAM (and some other nucleotide)
2.1
0.01
1.4
0.04
1.5
0.04



binding motif


CGI-117
Protein CGI-117
1.9
0.00
1.3
0.01
1.3
0.17


KDELR1
Required for the retention of luminal
1.9
0.04
1.5
0.02
1.3
0.20



endoplasmic reticulum proteins.


IFITM1
Implicated in the control of cell
1.6
0.02
1.4
0.01
1.3
0.24



growth.


COL7A1
Stratified squamous epithelial
1.5
0.05
1.4
0.02
1.9
0.31



basement membrane protein that form



anchoring fibrils which may contribute



to epithelial basement membrane



organization and adherence by



interacting with extracellular matrix



(ECM) proteins such as type IV



collagen









The data in FIGS. 13 and 18 also indicate that SEQ ID NO: 7 has both overlapping and distinct activities compared to LL-37.


The overall data extrapolated from this study indicates that the efficacy of host defense peptides can be improved significantly and specifically in order to obtain novel therapeutics that not only have anti-inflammatory properties, but also can specifically modulate responses in critical pathways that are involved in host defenses in pathogenesis.


EXAMPLE 15
Mechanisms of Action of Human Host Defense Peptide LL-37

LL-37 is a human cationic host defense peptide that is an essential component of innate immunity. It is a multifimctional modulator of innate immune responses demonstrating an ability to modulate gene regulation in certain cells, to alter cytokine expression in macrophages, to demonstrate chemotactic activity for neutrophils, monocytes and T cells, and to neutralize the endotoxic effects of lipopolysaccharide (LPS). However, the underlying mechanisms determining these effects of LL-37 were not clear. The general aim of this example is to determine how LL-37 affects initial cell signaling and to link cell signaling to some of these observed biological functions that are described herein such as the anti-endotoxin property and the ability to modulate cytokine and chemokine production in a human monocytic THP-1 cell line and human peripheral blood mononuclear cells (PBMCs).


To meet these goals, a variety of specific approaches were applied: (1) Assessing, by RT-PCR and ELISA, the ability of LL-37 to block the LPS-induced upregulation of transcription of certain genes and production of pro-inflammatory cytokines, respectively, in THP-1 cells and PBMCs; (2) Degradation of IκBα (known to be an NF-κB negative regulator) and translocation of the transcription factor NFκB were examined in the LPS-stimulated human THP-1 cell line in the presence of LL-37; (3) Since LPS and cytokine interleukin-1α (IL-1α) share similar signaling transduction pathways, it was interesting to compare the effects of LL-37 in modulating cytokine (IL-6) and chemokine (MCP-3) production in LPS- or IL-1α-stimualted human PBMCs; (4) Inhibitors (including oxidized ATP, an agonist of the P2X7 receptor, pertussis toxin, which inhibits G-protein coupled receptors, and LY294002, a PI3 Kinase inhibitor) were used to test if the synergistic effects of LL-37 were mediated by these receptors or pathways; (5) Western blots were performed to evaluate protein kinase B (Akt) phosphorylation and the activation of downstream transcription factors e.g., assessing the phosphorylation of cAMP-responsive element binding protein (CREB) and translocation of NFκB subunit p50 in human PBMCs. These analyses are designed to test if the PI3K-Akt-IκBα-NFκB and PI3K-Akt-CREB pathways are involved in LL-37-induced modulation of cytokine and chemokine release.


Methods


Western blotting—THP-1 cells (1×106 cells per condition) or PBMCs (5×106 cells per condition) were stimulated by adding LPS, LL-37 or endotoxin-free water as a vehicle control, and incubated for 20 min at 37° C., 5% CO2. After stimulation, the cells were centrifuged, washed once with ice-cold PBS with 1 mM vanadate and nuclear extracts were isolated using NE-PER Nuclear and Cytoplasmic Extraction Reagents Kit (Pierce) according to the manufacturer's instructions. The lysates were assayed for protein concentration using a BCA assay (Pierce). 7.5 μg nuclear lysate and 15 μg cytoplasm lysate were loaded per lane, resolved on a 8-10% SDS-PAGE at 120 V for 1 hr, and transferred to Immuno-blot PVDF membranes (Bio-Rad) for 75 min at 100 V. Immunoblot was performed using 1/1000 dilution of anti-p50, anti-total IκBα, anti-p-IκBα and anti-Akt (cell signaling) monoclonal antibodies in TBST/milk for 1 hour at room temperature. Membranes were washed for 30 min in TBST and then incubated with a 1/5000 dilution of HRP-conjugated goat anti-mouse or anti-rabbit Ab (in TBST/milk) for 1 hour. The membranes were washed for 30 min in TBST and developed with chemiluminescence peroxidase substrate (Sigma-Aldrich), according to manufacturer's instructions. The blots were reprobed with anti-total Akt or anti-GAPDH antibody as loading control.


Detection of cytokines—Fresh human PBMCs were plated at 8×105 cells in 1 ml of RPMI 1640 media (supplemented with 10% (v/v) heat-inactivated FBS, 1% (v/v) L-glutamine, 1 nM sodium pyruvate) respectively in 24 well plates. Cells were then incubated in media for periods of 24 hours in the presence of Pseudomonas aeruginosa PAO1 strain H103 LPS, LL-37, IL-1α (at the concentrations stated above), or endotoxin-free water as a vehicle control, in at least triplicate. Supernatants were collected and stored at −20° C. until use. The concentrations of IL-6 (eBioscience) and MCP-3 (R&D system) in the supernatants were measured using commercially prepared ELISA plates in accordance to the manufacturer's suggestion.


Results


LL-37 alone (20 μg/ml) caused IκBα degradation: protein levels of total IκBα diminished within 30 min and returned to control levels by 60 min in THP-1 cells, indicating that LL-37 may directly modulate elements of the LPS signaling pathway. Results are representative of three independent experiments (FIG. 19).


Fresh isolated human PBMCs were incubated with IL-1β (10 ng/ml) or LPS (100 ng/ml) in absence or presence of LL-37 (20 ug/ml) for 24 hours. IL-6 and MCP-3 ELISA were performed to measure the level of protein release (FIGS. 20A and 20B). For IL-6 production, stimulation with IL-1β resulted in an increase in IL-6 production, which could be significantly enhanced by the simultaneous addition of LL-37 even though LL-37 alone did not induce IL-6 release in THP-1 and PBMCs. In contrast, LL-37 inhibited LPS-triggered IL-6 release. Similar to IL-6, synergistic effects between IL-1β and LL-37 were also demonstrated for MCP-3 production. More importantly, LL-37 dramatically increased release of the MCP-3 in the presence of LPS, while LPS alone induced only low, but appreciable levels of MCP-3. Results are representative of 6 donors.


Cells were incubated with LL-37 alone or IL-1β (10 ng/ml) with or without LL-37 (20 μg/ml). Western blots were performed for cytoplasm protein and nuclear protein, showing that combined treatment of LL-37 and IL-1β showed higher IκBα phosphorylation after 30 min and p50 nuclear translocation after 60 min than LL-37 or IL-1β treatment alone in human PBMCs (FIG. 21A). Similar translocation results were also observed in THP-1 cells at an earlier time point (20 min after treatment). In addition, LL-37 alone induced NFκB subunit p50 translocation in both human PBMCs and THP-1 cells (FIG. 21B). Results are representative of three independent experiments for THP-1 cells and three donors for human PBMCs.


The immunomodulatory effects of LL-37 have been proposed to be dependent on signaling through a number of receptors, including the G-protein coupled receptor FPRL-1 and the purinergic receptor P2X7. Human PBMCs were pre-incubated with pertussis toxin (PTx, a G-protein coupled receptor inhibitor), oxidised ATP (OATP, a P2X7 receptor inhibitor) before exposure to LL-37 with or without IL-1β. MCP-3 production that resulted from combined treatment with LL-37 and IL-1β was dramatically inhibited by pre-treatment of PTx and partially blocked by oATP preincubation.


Since activated G-protein coupled receptor can lead to the elevated signaling of phosphatidylinositol 3-kinase (PI3 kinase) pathway, the role of PI3 kinase signaling in the above effects was further examined. Human PBMCs were pre-treated with PI3 kinase inhibitor, LY294002 (25 μM) for lh and following incubation with IL-1β (10 ng/ml) in presence or absence of LL-37 (20 μg/ml) for 24 hours (FIGS. 22A-22D). The LL-37 plus IL-1β mediated production of IL-6 and MCP-3 was significantly inhibited by LY294002 pre-incubation, indicating that PI3 kinase plays a role in LL-37-induced modulation of cytokine and chemokine production. Results are two representatives of three donors for human PBMCs.


Activation of PI3 kinase causes activation of a number of intracellular signal transduction pathways, including phosphorylation of the downstream target protein kinase B (Akt). Cytoplasm protein was isolated from human PBMCs treated with LL-37 with or without IL-1β. Western blot analysis showed that phosphorylation of Akt was observed in human PBMCs after exposure to LL-37 for 30 min (FIG. 23, top panel). Furthermore, activation of Akt was augmented by the presence of IL-1β (10 ng/ml). Results are representatives of three donors for human PBMCs.


The transcription factor CREB is downstream of the PI3 kinase→Akt pathway that leads to phosphorylation and activation of CREB and resulting in transcriptional up-regulation of a broad array of cellular responses. Western blot analysis showed that phosphorylation of CREB was observed in human PBMCs after exposure to LL-37 for 30 min and 60 min (FIG. 23, bottom panel). Furthermore, activation of Akt and CREB was augmented by the presence of IL-1β (10 ng/ml).


Although the invention has been described with reference to the presently preferred embodiment, it should be understood that various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims.

Claims
  • 1. A method of identifying an agent that is capable of selectively enhancing innate immunity comprising contacting a cell containing one or more genes that encode a polypeptide involved in innate immunity and protection against an infection, with an agent of interest, wherein expression of the one or more genes in the presence of the agent is modulated as compared with expression of the one or more genes in the absence of the agent, and wherein the modulated expression results in enhancement of innate immunity.
  • 2. An agent identified by the method of claim 1.
  • 3. The method of claim 1, wherein the agent does not stimulate a septic reaction.
  • 4. The method of claim 3, wherein the agent stimulates expression of the one or more genes, thereby selectively enhancing innate immunity.
  • 5. The method of claim 4, wherein the one or more genes are any gene shown in Table 69.
  • 6. The method of claim 4, wherein the one or more genes encode G-coupled protein receptors that initiate signaling from extracellular ligands.
  • 7. The method of claim 6, wherein the one or more genes are selected from the group consisting of GPR55, GPR6, GPR30, GPCR42, CASR, and EDG2.
  • 8. The method of claim 4, wherein the one or more genes encode chemokines or interleukins that attract immune cells.
  • 9. The method of claim 8, wherein one or more genes are delected from the group consisting of MCP-1, MCP-3, IL-8, CXCL-1, IL-17C, and IL-19.
  • 10. The method of claim 4, wherein the one or more genes encode receptors for chemokines.
  • 11. The method of claim 10, wherein the gene is CCR7.
  • 12. The method of claim 4, wherein the one or more genes encode transcription factors that mediate selective gene expression.
  • 13. The method of claim 12, wherein the one or more genes are selected from the group consisting of JAK1, STAT1, ELF1, Q9Y4C1, ETV4, POU1F1, ZNF254, ZNF292, ZNF78L1, HOXD3, and DLX5.
  • 14. The method of claim 4, wherein the one or more genes encode tyrosine-protein kinase or tyrosine-protein kinase receptors.
  • 15. The method of claim 14, wherein the one or more genes are selected from the group consisting of MAP2K6, NTRK3, PLCG1, EFNA2, and NCK1.
  • 16. The method of claim 4, wherein the one or more genes encode adhesion molecules that mediate cell attachment and interaction.
  • 17. The method of claim 16, wherein the one or more genes encode adhesion molecules of the ICAM, NCAM families, and PTPRF.
  • 18. The method of claim 17, wherein the one or more genes are selected from the group consisting of ICAM3, NCAM2, and PTPRF.
  • 19. The method of claim 4, wherein the one or more genes are involved in actin polymerization or cytoskeletal remodeling.
  • 20. The method of claim 19, wherein the one or more genes are selected from the group consisting of Integrin-α, EPHA4, ARHGAP6, and DST.
  • 21. The method of claim 4, wherein the one or more genes encode regulators of transcription factors.
  • 22. The method of claim 21, wherein the one or more genes are selected from the group consisting of TRIP4, GMEB2, GSK3B, ARNT, BACH1, ARID3A, HIPK2, POLR2D, TGIF, SSBP3, and FYB.
  • 23. The method of claim 4, wherein the one or more genes encode transmembrane receptors and adapters of signaling pathways.
  • 24. The method of claim 23, wherein the one or more genes are selected from the group consisting of WNT5B, FZD 10, TIRAP, and REPS 1.
  • 25. The method of claim 4, wherein the one or more genes encode proteins involved in antiviral activity.
  • 26. The method of claim 25, wherein the one or more genes are selected from the group consisting of IFNA2, STAT1, MNDA, and IFNA2.
  • 27. The method of claim 4, wherein the agent stimulates the JAK-STAT pathway.
  • 28. The method of claim 27, wherein the agent stimulates expression of one or more genes selected from the group consisting of JAK2, STAT1, STAT3, SOCS 1, and IL-19.
  • 29. The method of claim 4, wherein the agent stimulates the P13K pathway.
  • 30. The method of claim 29, wherein the agent stimulates expression of one or more genes selected from the group consisting of BACH2/PIK3CB, Akt, CREB, IL-6, and MCP-3.
  • 31. The method of claim 4, wherein the agent stimulates the ERK1/2 mitogen activated kinase pathway.
  • 32. The method of claim 31, wherein the agent stimulates expression of one or more genes selected from the group consisting of MAP3K1 and PP2A
  • 33. The method of claim 4, wherein the agent stimulates the p38 mitogen activated kinase pathway.
  • 34. The method of claim 33, wherein the agent stimulates expression of one or more genes selected from the group consisting of MINK1/MAP4K6, MAP2K6, and MAP2K4.
  • 35. The method of claim 4, wherein the agent transiently stimulates the NFYB pathway.
  • 36. The method of claim 35, wherein the agent stimulates expression of one or more genes selected from the group consisting of TIRAP, NFκB2 (p52), DUSP14, ICAM3, TRIP4, MMP17, ITGB4, ZNF36, ZNF251, BNIP1, CD226, NRXN1, and TNC.
  • 37. The method of claim 4, wherein the agent stimulates the AP-1, JNK or Wnt pathways.
  • 38. The method of claim 37, wherein the agent stimulates expression of one or more genes selected from the group consisting of TRIP4, TIRAP, HIPK2, GSK3B, and FZD10.
  • 39. A method of identifying a pattern of gene expression for identification of an agent that selectively enhances innate immunity comprising: contacting a cell containing one. or more genes that encode a polypeptide involved in innate immunity and defense against infections, with an agent of interest, wherein expression of the one or more genes in the presence of the agent is modulated as compared with expression of the one or more genes in the absence of the agent, and wherein the modulated expression results in. enhancement of innate immunity.
  • 40. The method of claim 39, wherein the modulated expression is a marker of enhancement of innate immunity.
  • 41. The method of claim 40, further comprising determining the efficacy of compounds that enhance innate immunity.
  • 42. The method of claim 40, wherein the one or more genes are any gene shown in Table 69.
  • 43. The method of claim 42, wherein the one or more genes express IL-8, IL-6, IL-19, CXCL-1, MCP-3, or MCP-1.
  • 44. The method of claim 39, wherein the modulated expression occurs in the presence of a bacterial signature molecule.
  • 45. The method of claim 44, wherein the bacterial signature molecule is a Toll-like receptor agonist.
  • 46. The method of claim 45, wherein the Toll-like receptor agonist is selected from the group consisting of bacterial lipopolysaccharide, lipoteichoic acid, and CpG bacterial signature DNA.
  • 47. The method of claim 44, wherein the one or more genes are any gene shown in Table 71.
  • 48. A method of identifying an agent that is capable of selectively enhancing innate immunity in the presence of an infection or bacterial signature molecule comprising: contacting a cell containing one or more genes that encode a polypeptide involved in innate immunity, with an agent of interest in the presence of a bacterial signature molecule, wherein expression of the one or more genes in the presence of the agent and bacterial signature molecule is modulated as compared with expression of the one or more genes in the absence of the agent and bacterial signature molecule, and wherein the modulated expression results in enhancement of innate immunity.
  • 49. An agent identified by the method of claim 48.
  • 50. The method of claim 48, wherein the bacterial signature molecule is a Toll-like receptor agonist.
  • 51. The method of claim 50, wherein the Toll-like receptor agonist is selected from the group consisting of bacterial lipopolysaccharide, lipoteichoic acid, and CpG bacterial signature DNA.
  • 52. The method of claim 51, wherein the one or genes are any gene shown in Table 71.
  • 53. The method of claim 52, wherein the agent does not stimulate a septic reaction.
  • 54. The method of claim 48, wherein the agent has anti-endotoxic activity.
  • 55. The method of claim 54, wherein the one ore more genes are selected from the group consisting of GPD1, Q8NI35, FEZ2, NRXN1, PLCG1, Q7RTU0, ALDOB, Q9H5P1, SYT11, UBXD2, PROZ, PLAC8, Q96PN6, ASTN2, O60290, FTCD, NFKB2, CTLA4, PSMA 1, CCL2, HNF4A, MAFF, FBXO32, TNFα, NPAS2, ICAM3, Q8NC30, Q8IUC6, O94940, CGI-117, KDELR1, IFITM1 and COL7A1.
  • 56. The method of claim 48, wherein the agent stimulates transient IKcBa degradation or transient NFκB subunit p50 translocation.
  • 57. The method of claim 48, further comprising contacting the cell with IL-1β.
  • 58. The method of claim 48, wherein the one-or more genes encode chemokines.
  • 59. The method of claim 58, wherein the one or more genes are selected from the group consisting of CCL20, CCL23, IL-6, and MCP-3.
  • 60. The method of claim 48, wherein the one or more genes encode cytokine receptors.
  • 61. The method of claim 60, wherein the one or more genes are EBI3 or IL7R.
  • 62. The method of claim 48, wherein the one or more genes encode factors involved in lymphocyte activation.
  • 63. The method of claim 62, wherein the one or more genes are selected from the group consisting of SLAMF1, CD58, and IL32.
  • 64. The method of claim 48, wherein the one or more genes encode regulators of signal transduction.
  • 65. The method of claim 64, wherein the one or more genes are selected from the group consisting of MAP2K2, DUSP5, MAPK8IP3, RIN2, RANBP9, IP3 3-kinase A, BATF, IRAK3, NM1, SP3, RAP2C, PNRC1, NEK1, CHC1, ZNF219, ZNF593, WIF1, PIM2, CD79A, and LATS2.
  • 66. The method of claim 48, wherein the one or more genes encode substrate transporters.
  • 67. The method of claim 66, wherein the one or more genes are SLC23A3 or SLC17A5.
  • 68. The method of claim 48, wherein the one or more genes encode apoptosis regulators.
  • 69. The method of claim 68, wherein the one or more genes are selected from the group consisting of BOK, BIRC3, TNFRSF6, and CASP9.
  • 70. The method of claim 48, wherein the one or more genes encode genes associated with plasma membrane.
  • 71. The method of claim 70, wherein the one. or more genes are selected from the group consisting of STIM1, BPAG1, PTPN4, TRIM36, SDK1, and FNDC5.
  • 72. The method of claim 48, wherein the one or more genes encode genes involved in selective ion transport and in mediating selective ion-channels.
  • 73. The method of claim 72, wherein the one or more genes are selected from the group consisting of VGCNL1, TRPC5, CACNA1B, KCNA6, KCNJ2, KCNA10, and AQP9.
  • 74. The method of claim 48, wherein the one or more genes encode growth modulating genes or genes involved in wound healing.
  • 75. The method of claim 74, wherein the one or more genes are FGF10 or AREG.
  • 76. The method of claim 48, wherein the one or more genes encode inflammatory mediators.
  • 77. The method of claim 76, wherein the one or more genes are selected from the group consisting of PTGS2, SOD2, TNFAIP8, and TNIP3.
  • 78. The method of claim 48, further comprising contacting the cell with IL-1β, wherein the agent stimulates the PI3 kinase pathway.
  • 79. The method of claim 78, wherein the agent stimulates transient IκBα phosphorylation and p50 nuclear translocation.
  • 80. The method of claim 78, wherein the one or more genes encodes a G-protein coupled receptor or a purinergic receptor.
  • 81. The method of claim 78, wherein the purinergic receptor is P2X7.
  • 82. The method of claim 78, wherein the agent further stimulates phosphorylation of Akt.
  • 83. The method of claim 82, wherein the phosphorylation of Akt stimulates activation of CREB.
  • 84. A method of identifying an agent that selectively reduces inflammation comprising: contacting a cell containing one or more genes that encode a polypeptide involved in sepsis, with an agent of interest, wherein the agent reduces expression of the one or more genes compared with expression of the one or more genes in the absence of the agent.
  • 85. The method of claim 84, wherein the one or more genes are selected from the group consisting of GPD1, Q8NI35, FEZ2, NRXN, PLCG1, Q7RTU0, ALDOB, Q9H5P1, SYTI 11, UBXD2, PROZ, PLAC8, Q96PN6, ASTN2, O60290, FTCD, NFKB2, CTLA4, PSMA1, CCL2, HNF4A, MAFF, FBXO32, TNF, NPAS2, ICAM3, Q8NC30, Q81UC6, O94940, CGI-117, KDELR1, IFITM1, and COL7A1.
RELATED APPLICATION DATA

This application claims priority under 35 U.S.C. §120 to U.S. patent application Ser. No. 11/241,882, filed Sep. 29, 2005, which is a continuation-in-part of U.S. patent application Ser. No. 10/661,471, filed Sep. 12, 2003, which is a continuation-in-part of U.S. patent application Ser. No. 10/308,905, filed Dec. 2, 2002, which claims priority under 35 U.S.C. §119(e) to U.S. patent application Ser. No. 60/336,632, filed Dec. 3, 2001, herein incorporated by reference in their entirety.

Provisional Applications (1)
Number Date Country
60336632 Dec 2001 US
Continuation in Parts (3)
Number Date Country
Parent 11241882 Sep 2005 US
Child 11400411 Apr 2006 US
Parent 10661471 Sep 2003 US
Child 11241882 Sep 2005 US
Parent 10308905 Dec 2002 US
Child 10661471 Sep 2003 US