Antimicrobial Peptides

Information

  • Patent Application
  • 20230086360
  • Publication Number
    20230086360
  • Date Filed
    September 14, 2022
    a year ago
  • Date Published
    March 23, 2023
    a year ago
Abstract
Provided herein are synthetic peptides with enhanced antimicrobial and antibiofilm characteristics, and are biocompatible with mammalian cellular systems. The disclosed synthetic antimicrobial moieties include a mastoparan peptide having SEQ ID NO:1 and a pentapeptide motif formed from phenylalanine, leucine, proline, and two isoleucine residues, wherein the pentapeptide motif is conjugated the N-terminus of the mastoparan peptide. Also provided are compositions comprising the synthetic peptides, as well as methods of treating a microbial infection or removing a biofilm using the peptides.
Description
SEQUENCE LISTING

The instant application contains a sequence listing that has been submitted electronically in .XML format and is hereby incorporated by reference in its entirety. Said .XML copy, created on Sep. 14, 2022, is named 103241006873.xml and is 108,256 bytes in size.


TECHNICAL FIELD

The present disclosure pertains to peptides with antimicrobial properties.


BACKGROUND

The excessive use and misuse of antibiotics has led to increasing rates of resistance in pathogenic bacteria(1). According to the Centers for Disease Control and Prevention (CDC) report in 2019, 2.8 million antibiotic-resistant infections occur in the US each year, leading to approximately 35,000 deaths annually(2). In addition, 19.7% (-11 million) of all global deaths in 2017 were caused by sepsis, a syndrome most commonly linked to bacterial infections and for which antibiotics are the primary treatment(3). Drug-resistant infections are not only serious on their own. Secondary infections are also highly relevant and represent a major cause of death in global pandemics, being responsible for 95% of deaths during the 1918 flu pandemic(4) and also playing a role in the COVID-19 pandemic(5).


The emergence of multidrug-resistant bacterial pathogens has coincided with a severe decline in the development and approval of new antibacterial drugs(1). Thus, there is an urgent need for novel antibiotics effective against drug-resistant bacteria.


SUMMARY

Disclosed herein are antimicrobial peptides that comprise a mastoparan peptide having SEQ ID NO:1 and a pentapeptide motif formed from phenylalanine, leucine, proline, and two isoleucine residues, wherein the pentapeptide motif is conjugated the N-terminus of the mastoparan peptide.


Also provided are methods of treating a microbial infection in a subject comprising administering to the subject a therapeutically effective amount of an antimicrobial peptide according to the present disclosure.


The present disclosure also provides methods of removing a biofilm comprising contacting the biofilm with an effective amount of an antimicrobial peptide according to the present disclosure.


The present disclosure also pertains to methods of forming an antimicrobial peptide comprising conjugating a pentapeptide motif formed from phenylalanine, leucine, proline, and two isoleucine residues to a mastoparan peptide having SEQ ID NO:1 at the N-terminus of the peptide.





BRIEF DESCRIPTION OF THE DRAWINGS

The file of this patent or application contains at least one drawing/photograph executed in color. Copies of this patent or patent application publication with color drawings/photographs will be provided by the Office upon request and payment of the necessary fee.



FIG. 1A provides a schematic representation of the design approach based on FIG. 1B the insertion into the mast-L sequence of a pentapeptide motif (FLPII) conserved in naturally occurring peptides (e.g., brevenin, temporin and vespid chemotactic peptide families) with potent antimicrobial, immunomodulatory and chemotactic properties. Such insertion yielded the synthetic peptide mast-MO.



FIGS. 2A-2C pertains to analyses of the natural wasp venom toxin and its synthetic derivative. As provided in FIG. 2A, circular dichroism experiments showed that mast-L and mast-MO presented helical patterns in all tested media: DPC (15 mmol L-1), SDS (100 mmol L-1), TFE/Water (1/1, v/v), and PCPG (1 mmol L-1). The a-helical structures were confirmed by nuclear magnetic resonance. The TOCSY contour maps for mast-MO, as depicted in FIG. 2B, where the fingerprint region in H20/D20 (9:1, v/v) indicate a helical pattern. In FIG. 2C, the hydrogen by deuterium exchange experiment (H/D exchange) shows the orientation of mast-MO on the surface of SDS-d25 micelles. The slow H/D exchange demonstrated the high affinity between mast-MO and SDS micelles. Residues Ile4, Ala10, Leull, Leu14, Lys17, Ile18 did not exchange the amide hydrogen for deuterium, because these amide groups are compromised interacting with the micelles. After seven days, residues Leu14 and Lys17 had not yet undergone H/D exchange, indicating that both residues may be in contact with the micelle or forming resistant hydrogen bonds thus preventing the H/D exchange (PDB ID: mastoparan-L - 6DUL - and mastoparan-MO - 6DUU).



FIG. 3A depicts the antimicrobial activity of mast-L, mast-MO and standard-of-care antibiotics gentamicin and imipenem against 15 clinically relevant pathogens. FIG. 3B shows the cytoplasmic membrane depolarization effects of mast-MO against 8 different Gram-negative pathogens determined using the DiSC3(5) assay and FIG. 3C provides an NPN assay demonstrating the ability of mast-MO to potently damage and permeabilize the bacterial outer membrane compared to the controls in the absence of mast-MO.



FIG. 4A provides a schematic of different interactions displayed by mastoparan peptides on the surface of biological membranes leading to internalization into the cell and subsequent immunomodulatory effects. As shown in FIG. 4B, mast-MO (10 mg Kg′) triggered leukocyte recruitment to the E. coli ATCC8739 infection site to levels comparable to the positive control TGA, FIG. 4C shows release over time (48 h) of IL-12 and FIG. 4D depicts release over 48 h of TNF-α into the peritoneal cavity of mice. Cytokines monitored in mice for 24 h after infection with (as shown in FIG. 4E) E. coli ATCC8739 and (as shown in FIG. 4F) S. aureus ATCC29213. Cytokine release (i.e., IFN-γ, IL-6, IL-10, IL-12p70 and TNF-α) into the peritoneal cavity of C57BL/6 mice was detected and quantified 24 h after infection with E. coli ATCC8739 and S. aureus ATCC29213. In all experiments, mice were treated with 10 mg Kg−1 of mastoparan-MO and 5 mg Kg−1 of mastoparan-L. PBS and imipenem (10 mg Kg−1) were used as negative and positive controls, respectively. Data were expressed as mean±standard deviation. Statistical analysis was performed using Bonferroni test. *** p<0.001 as significant compared to the control.



FIG. 5A-5I pertains to a study of the anti-infective activity of mast-MO against Gram-negative and Gram-positive pathogens in a lethal sepsis infection model. In FIG. 5A mice were infected with bacteria, treated with each peptide or antibiotic and monitored for 8 days. Survival and cell counts after 24 h, respectively, are shown for E. coli ATCC8739 (2×104 CFU/mL inoculum) FIG. 5B and FIG. 5C, S. aureus ATCC29213 (2×109CFU/mL inoculum) FIG. 5D and FIG. 5E, E. coli 1812446 (KPC-positive) (2×104 CFU inoculum) FIG. 5F and FIG. 5G, and S. aureus (MRSA) ATCC33591 (2×109 CFU inoculum) FIG. 5H and FIG. 51. Mice were treated intraperitoneally (i.p.) with 1, 5 and 10 mg Kg−1 of mast-L, mast-MO or 10 mg Kg−1 of the antibiotics gentamicin and imipenem. PBS was used as a control for the untreated group of mice. Treatments were administered 3 h post-infection. Data are expressed as the mean±standard deviation. Statistical analysis was performed using Bonferroni test. * p<0.1, ** p<0.01, *** p<0.001 compared to the untreated control.



FIG. 6A-6F pertain to a study demonstrating that permutations of the conserved motif lead to mast-MO variants that lack toxicity against human cells and display anti-infective activity in a mouse model. FIG. 6A provides a schematic showing the dual immunomodulatory and antimicrobial activities of mast-MO demonstrated in this work. The peptide killed bacterial cells by destabilizing their outer membrane and displayed immunomodulatory properties by attracting leukocytes to the site of infection and anti-inflammatory activity by repressing pro-inflammatory mediators (e.g., TNF-α and IL-12). Despite these therapeutic properties, mast-MO retained some cytotoxicity towards human cells at concentrations immediately above its antimicrobial and immunomodulatory activities hindering its use as a potential therapy in humans. FIG. 6B pertains to design of mast-MO derivatives through permutation of its FLPII motif. FIG. 6C depicts cytotoxic activity of mast-MO and its permutation variants against human embryonic kidney cells (i.e., HEK293).



FIG. 6D provides a schematic of the experimental design. Briefly, the back of mice was shaved, and an abrasion was generated to damage the stratum corneum and the upper layer of the epidermis. Subsequently, an aliquot of 50 μL containing 5×107 CFU/20 μL of P. aeruginosa in PBS was inoculated over each defined area. One day post-infection, peptides (16 μmol L−1) were administered to the infected area. In FIG. 6E, mouse body weight measurements throughout the experiment were taken and normalized by the body weight of non-infected mice as a proxy for potential toxicity. FIG. 6F illustrates the anti-infective activity of the permutation-based synthetic analogs compared to mast-MO-treated and negative control groups. Briefly, mice were anesthetized with isoflurane, had their backs shaved and a superficial linear skin abrasion was made with a needle in order to damage the stratum corneum and upper layer of the epidermis. A bacterial load 1×107 CFU/20 μL of bacteria (in 20 μL) in PBS was inoculated over each defined area containing the scratch with a pipette tip. One day after the infection, a single dose of each peptide (16 μmol L−1) was administered to the infected area. Six animals per group were euthanized and the area of scarified skin was excised two days post-infection, homogenized using a bead beater for 20 min (25 Hz), and serially diluted for CFU quantification (statistical significance was determined using two-way ANOVA followed by Dunnett's test, ***p<0.001).



FIG. 7 illustrates the antimicrobial activity of Mast-MO against clinically relevant Gram-negative bacterial strains. Two-fold dilutions of peptide ranging from 0 to 128 wino′ L-1 in LB medium were added to 96-wells and exposed to 105 bacterial cells and incubated for 24 h at 37° C. The optical density at 600 nm (OD600) was measured in a microplate reader after 24 h (red meaning very low or absent growth and blue meaning growth of bacterial cells). Experiments were performed in three biological replicates.



FIGS. 8A and 8B illustrate the anti-biofilm activity of mast-MO. FIG. 8A depicts how most bacterial infections are present as surface-associated communities called biofilms. Biofilms are aggregates of microorganisms in which cells are embedded in a self-produced matrix of extracellular polymeric substrates that adhere to other aggregates or onto a surface(49). The biofilm state leads to overall increased antibiotic resistance profiles(50). Therefore, we tested whether mast-MO displayed antibiofilm activity using a dynamic flow cell apparatus as previously described(12, 51, 52). Glassy flow cells were colonized with S. aureus ATCC33591 and E. coli ATCC0157, and after 48 h were exposed to the peptide treatment or no treatment (untreated control). After 72 h, the medium flow was stopped, and cells were stained with SYTO-9 and propidium iodide (PI). Cell membranes were stained in green and dead cells were stained in red. Untreated flow cells colonized by (FIG. 8B) S. aureus and (FIG. 8E) E. coli showed slightly higher number of cells than the ones observed in the treated flow cells with mast-L as shown in FIG. 8C and FIG. 8F for S. aureus and E. coli, respectively. Mast-MO, on the other hand, showed high inhibition of biofilm formation, inhibiting almost completely the adhesion of cells to the glassy surface of the flow cells, FIG. 8D and FIG. 8G for S. aureus and E. coli, respectively.



FIG. 9 depicts the results of bacterial outer membrane permeabilization assays using the hydrophobic dye NPN. Briefly, the membrane permeability of each bacteria exposed to mast-MO (20 wino′ L−1) was determined by using the N-phenyl-1-napthylamine (NPN) uptake assay. Bacteria were grown to OD600 of 0.4, centrifuged (10,000 rpm at 4° C. for 10 min), washed and resuspended in buffer (5 mmol L−1 HEPES, 5 mmol L−1 glucose, pH 7.4). Four 4 of NPN solution (0.5 mmol L−1— working concentration of 10 μmol L−1 after dilutions) was added to 1004 of bacterial solution in white 96-well plates. The background fluorescence was recorded at Xex=350 nm and Xem=420 nm. Mast-MO samples in water (1004 solution at 1.2 mol L−1) were added to the 96-wells plate, and fluorescence was recorded as a function of time until no further increase in fluorescence was observed (at 20 min). Mast-MO was able to disturb the outer membrane of all bacteria tested by permeabilization.



FIG. 10 depicts colony forming units (CFU) count assays to quantify the antimicrobial activity of mast-MO and its synthetic analogs. Briefly, 105 bacterial cells and serially diluted (0-64 μmoL−1) peptides were added to a 96-well plate and incubated at 37° C. One day after exposing bacteria to the peptides, the solution in each well was 10-fold diluted seven times and the serial dilutions were plated in agar plates, which were incubated for 22 h at 37° C. (B) Next, bacterial colonies were counted. All assays were performed in three biological replicates (statistical significance was determined using two-way ANOVA followed by Dunnett's test, error bars=standard error of the mean, ns=statistically not significant, *p<0.05, “p<0.005, ***p<0.001, ****p<0.0001).





DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS

The presently disclosed inventive subject matter may be understood more readily by reference to the following detailed description taken in connection with the accompanying examples, which form a part of this disclosure. It is to be understood that these inventions are not limited to the specific formulations, methods, articles, or parameters described and/or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting of the claimed inventions.


The entire disclosures of each patent, patent application, and publication cited or described in this document are hereby incorporated herein by reference.


As employed above and throughout the disclosure, the following terms and abbreviations, unless otherwise indicated, shall be understood to have the following meanings.


In the present disclosure the singular forms “a,” “an,” and “the” include the plural reference, and reference to a particular numerical value includes at least that particular value, unless the context clearly indicates otherwise. Thus, for example, a reference to “an microbe” is a reference to one or more of such organisms and equivalents thereof known to those skilled in the art, and so forth. Furthermore, when indicating that a certain element “may be” X, Y, or Z, it is not intended by such usage to exclude in all instances other choices for the element.


When values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. As used herein, “about X” (where X is a numerical value) preferably refers to ±10% of the recited value, inclusive. For example, the phrase “about 8” can refer to a value of 7.2 to 8.8, inclusive. This value may include “exactly 8”. Where present, all ranges are inclusive and combinable. For example, when a range of “1 to 5” is recited, the recited range should be construed as optionally including ranges “1 to 4”, “1 to 3”, “1-2”, “1-2 & 4-5”, “1-3 & 5”, and the like. In addition, when a list of alternatives is positively provided, such a listing can also include embodiments where any of the alternatives may be excluded. For example, when a range of “1 to 5” is described, such a description can support situations whereby any of 1, 2, 3, 4, or 5 are excluded; thus, a recitation of “1 to 5” may support “1 and 3-5, but not 2”, or simply “wherein 2 is not included.”


In the present disclosure, numerals appearing in parentheses refer to the correspondingly numbered references that appear infra under the heading “References”.


Toxins represent a previously untapped source of potential antimicrobials. They are contained in venoms and have evolved (e.g., in plants, animals and microbes) as part of defensive strategies. Typically, crude venoms contain a wide range of different peptide toxins, many of which are small in size, easy to synthesize, structurally stable, and highly biodiverse(6). Antimicrobial peptides (AMPs), a promising class of antibiotic candidates, have been found in venoms(7). AMPs have an almost unlimited sequence space and multiple mechanisms of action against microorganisms(1, 7-13). These agents can present broad- or narrow-spectrum activity, and rapid antimicrobial(7, 9, 10, 14) and immunomodulatory(l1, 15-17) properties, which allow them to act against a range of microorganisms, including Gram-negative and Gram-positive bacteria, fungi, viruses, and parasites. Relevant to their potential clinical translatability, bacteria exposed to AMPs have been shown to develop resistance at a much lower rate than when exposed to conventional antibiotics (18).


Cationic peptides displaying an a-helical structure constitute a class of AMPs found in venoms. This class consists of small amphipathic peptides with net positive charge that tend to adopt an a-helical structure upon contact with hydrophilic/hydrophobic interfaces, such as the membrane of microorganisms(8). One such example is mast-L, an AMP from the mastoparan family, derived from the venom of the social wasp Vespula lewisii(19). The amino acid sequence of mast-L is NLKALAALAKKIL (SEQ ID NO:1). However, the use of this peptide as a therapeutic has been hindered by its toxicity profile, as it displays high hemolytic activity(l3) and promotes massive mast cell degranulation(20, 21).


The present inventors have introduced modifications to mast-L in order to increase the therapeutic properties of the protein, while decreasing its inherent cytotoxicity. Through the approach disclosed herein, the inventors have generated novel anti-infective synthetic peptides that lack toxicity and display potent antimicrobial and immunomodulatory activities in clinically relevant sepsis and skin infection animal models.


Disclosed herein are antimicrobial peptides that comprise a mastoparan peptide having SEQ ID NO:1 and a pentapeptide motif formed from phenylalanine, leucine, proline, and two isoleucine residues, wherein the pentapeptide motif is conjugated the N-terminus of the mastoparan peptide.


Multidrug-resistant pathogens are becoming increasingly prevalent and no new classes of antibiotics have been discovered for decades. Therefore, strategies enabling the discovery of new strategies for treating infections are urgently needed. Toxins from the venom of insects and arthropods are rich in antimicrobials that represent an untapped source of template molecules for the design of novel drug candidates. The present inventors have leveraged a design strategy to convert a highly toxic peptide derived from wasp venom, namely, the mast-L protein from Vespula lewisii, into non-toxic synthetic derivatives with drug-like properties and anti-infective activity in pre-clinical animal models. Initially, a pentapeptide motif highly conserved in existing chemotactic and antimicrobial peptides was engineered into mast-L. In relation to mast-L, the resulting synthetic peptide, dubbed mast-MO, presented enhanced antimicrobial activity by destabilizing the bacterial outer membrane, and exhibited immunomodulatory properties by increasing leukocyte migration to the infection site and repressing pro-inflammatory factors, crucial to allow infection clearance. In animal models, mast-MO treatment caused a decrease in pro-inflammatory cytokines IL-12, TNF- a and IL-6 at the site of infection and increased leukocyte migration required for infection resolution. Since mast-MO retained some toxicity against human cells, the inventors also built, via permutation design, a second generation of analogs (FIG. 6). Seven identified synthetic peptides were no longer toxic against human cells and presented increased anti-infective activity against clinically relevant bacteria both in vitro and in vivo (FIG. 6).


The dual antimicrobial and immunomodulatory actions of mast-MO enable it to directly kill bacterial pathogens and selectively enhance host immune responses to clear infections while maintaining control of the inflammatory response. Multifunctional antimicrobials such as the ones presented here represent a new paradigm for treating infections by targeting both bacteria and the host. Moreover, the peptide did not induce any apparent immunotoxicities in any of the mouse models tested.


In some embodiments, the antimicrobial peptide comprises any one of SEQ ID NOS:5-64, as provided in Table 7, infra. In particular embodiments, the antimicrobial peptide comprises SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:31, SEQ ID NO:38, SEQ ID NO:52, SEQ ID NO:54, or SEQ ID NO:59.


The present disclosure also provides compositions for treating a microbial infection comprising a therapeutically effective amount of an antimicrobial peptide according to any one of the embodiments described herein. Also provided herein are methods of treating a microbial infection in a subject comprising administering to the subject a therapeutically effective amount of an antimicrobial peptide according to the present disclosure. As described above, the present inventors have discovered that the antimicrobial peptides disclosed herein possess antimicrobial characteristics, with better biocompatibility than the mast-L peptide, and therefore represent alternatives both to traditional antibiotic compounds to which microbial resistance has arisen or is likely to arise, and to naturally occurring AMPs that possess unacceptably high levels of toxicity to mammalian cells.


As used herein, the phrase “therapeutically effective amount” refers to the amount of active agent (here, the antimicrobial peptide) that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following:


(1) at least partially preventing the disease or condition or a symptom thereof; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease;


(2) inhibiting the disease or condition; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., including arresting further development of the pathology and/or symptomatology); and


(3) at least partially ameliorating the disease or condition; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., including reversing the pathology and/or symptomatology).


The antimicrobial peptides according to the present disclosure may be provided in a composition that is formulated for any type of administration. For example, the compositions may be formulated for administration orally, topically, parenterally, enterally, or by inhalation (e.g., intranasally). The active agent may be formulated for neat administration, or in combination with conventional pharmaceutical carriers, diluents, or excipients, which may be liquid or solid. The applicable solid carrier, diluent, or excipient may function as, among other things, a binder, disintegrant, filler, lubricant, glidant, compression aid, processing aid, color, sweetener, preservative, suspensing/dispersing agent, tablet-disintegrating agent, encapsulating material, film former or coating, flavoring agent, or printing ink. Any material used in preparing any dosage unit form is preferably pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active agent may be incorporated into sustained-release preparations and formulations. Administration in this respect includes administration by, inter alia, the following routes: intravenous, intramuscular, subcutaneous, intraocular, intrasynovial, transepithelial including transdermal, ophthalmic, sublingual and buccal; topically including ophthalmic, dermal, ocular, rectal and nasal inhalation via insufflation, aerosol, and rectal systemic.


In powders, the carrier, diluent, or excipient may be a finely divided solid that is in admixture with the finely divided active ingredient. In tablets, the active ingredient is mixed with a carrier, diluent or excipient having the necessary compression properties in suitable proportions and compacted in the shape and size desired. For oral therapeutic administration, the active compound may be incorporated with the carrier, diluent, or excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. The amount of active agent(s) in such therapeutically useful compositions is preferably such that a suitable dosage will be obtained.


Liquid carriers, diluents, or excipients may be used in preparing solutions, suspensions, emulsions, syrups, elixirs, and the like. The active ingredient of this invention can be dissolved or suspended in a pharmaceutically acceptable liquid such as water, an organic solvent, a mixture of both, or pharmaceutically acceptable oils or fat. The liquid carrier, excipient, or diluent can contain other suitable pharmaceutical additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers, or osmo-regulators.


Suitable solid carriers, diluents, and excipients may include, for example, calcium phosphate, silicon dioxide, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, ethylcellulose, sodium carboxymethyl cellulose, microcrystalline cellulose, polyvinylpyrrolidine, low melting waxes, ion exchange resins, croscarmellose carbon, acacia, pregelatinized starch, crospovidone, HPMC, povidone, titanium dioxide, polycrystalline cellulose, aluminum methahydroxide, agar-agar, tragacanth, or mixtures thereof


Suitable examples of liquid carriers, diluents and excipients, for example, for oral, topical, or parenteral administration, include water (particularly containing additives as above, e.g. cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g. glycols) and their derivatives, and oils (e.g. fractionated coconut oil and arachis oil), or mixtures thereof


For parenteral administration, the carrier, diluent, or excipient can also be an oily ester such as ethyl oleate and isopropyl myristate. Also contemplated are sterile liquid carriers, diluents, or excipients, which are used in sterile liquid form compositions for parenteral administration. Solutions of the active agents can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. A dispersion can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.


The pharmaceutical forms suitable for injectable use include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form is preferably sterile and fluid to provide easy syringability. It is preferably stable under the conditions of manufacture and storage and is preferably preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier, diluent, or excipient may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of a dispersion, and by the use of surfactants. The prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In some instances, the antimicrobial peptides themselves may be sufficient to prevent contamination by microorganisms. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions may be achieved by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions may be prepared by incorporating the active agent in the pharmaceutically appropriate amounts, in the appropriate solvent, with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions may be prepared by incorporating the sterilized active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation may include vacuum drying and freeze drying techniques that yield a powder of the active ingredient or ingredients, plus any additional desired ingredient from the previously sterile-filtered solution thereof


Thus, an antimicrobial peptide may be in the present compositions and methods in an effective amount by any of the conventional techniques well-established in the medical field. For example, the administration may be in the amount of about 0.1 mg/day to about 500 mg per day. In some embodiments, the administration may be in the amount of about 250 mg/kg/day. Thus, administration may be in the amount of about 0.1 mg/day, about 0.5 mg/day, about 1.0 mg/day, about 5 mg/day, about 10 mg/day, about 20 mg/day, about 50 mg/day, about 100 mg/day, about 200 mg/day, about 250 mg/day, about 300 mg/day, or about 500 mg/day.


Also disclosed are methods comprising contacting a biofilm with an effective amount of an antimicrobial peptide according to the present disclosure. Such methods may be effective to remove or reduce the presence of an unwanted biofilm, such as in hospitals or other medical settings, in sewer and filtration systems, in industrial settings, on equipment involved in food preparation or manufacture, in aquaculture or hydroponics, or in any other context that is prone to unwanted biofilm formation.


In accordance with the methods of treating a microbial infection in a subject or the methods comprising contacting a biofilm according to the present disclosure, microbes against which the present antimicrobial peptides are effective may be, for example, any unicellular organism, such as gram-negative bacteria, gram-positive bacteria, protozoa, viruses, bacteriophages, and archaea. The present peptides can have an antimicrobial effect with respect to any such microbe. Examples of bacteria against which the present compounds are effective to cause reduction in numbers include gram positive bacteria and gram negative bacteria, for example, Salmonella enterica, Listeria monocytogenes, Escherichia coli, Clostridium botulinum, Clostridium difficile, Campylobacter, Bacillus cereus, Vibrio parahaemolyticus, Vibrio cholerae, Vibrio vulnificus, Staphylococcus aureus, Yersinia enterocolitica, Shigella, Moraxella spp., Helicobacter, Stenotrophomonas, Bdellovibrio, Legi onella spp. (e.g., pneumophila), Neisseria gonorrhoeae, Neisseria meningitidis, Haemophilus influenzae, Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, Proteus mirabilis, Enterobacter cloacae, Serratia marcescens, Helicobacter pylori, Salmonella enteritidis, Salmonella typhi, and combinations thereof. Examples of Salmonella enterica serovars that can be reduced using the compounds of the disclosure include, for example, Salmonella enteriditis, Salmonella typhimurium, Salmonella poona, Salmonella heidelberg, and Salmonella anatum. Exemplary viruses against which the present peptides are effective to cause reduction in numbers include coronaviruses, rhinoviruses, and influenza viruses.


Hereinafter, the present disclosure will be described in more detail through Examples, which are intended to be illustrative to the present disclosure, although present disclosure is not limited to the Examples.


Example 1— Generation of Novel Antimicrobial Peptides


Design and structural analyses of wasp venom derived peptides. Despite the promise of mast-L as an antimicrobial, its activity must be improved to surpass that of standard-of-care antibiotics in order to enter clinical trials. In order to increase its antimicrobial and immunomodulatory properties, we performed a comprehensive search and analysis of the Antimicrobial Peptides Database (APD3)(22) looking for conserved motifs associated with such biological functions. The pentapeptide motif (FLPII) was identified to be conserved in the N-terminal extremity of peptides with high antimicrobial and immunomodulatory (e.g., chemotactic) activities, including brevenins(23, 24), temporins(24), vespid chemotactic peptides(25) (FIG. 1A and Table 1) and mediating interactions with membranes in plant lipocalins (26).









TABLE 1







Natural peptides with potent antimicrobial and


immunomodulatory activities containing the


conserved FLPII pentapeptide motif in their N-


terminal extremity.












SEQ





ID



Family
Peptide
NO
Sequence





Brevenins
Brevinin-1BLb
65

FLPIIAGVAAKVLPKIFCAISKKC




Brevinin-1BLc
66

FLPIIAGIAAKFLPKIFCTISKKC




Brevinin-1CHa
67

FLPIIAGVAAKVLPKLFCAITKKC




Brevinin-1Pa
68

FLPIIAGVAAKVFPKIFCAISKKC




Brevinin-1Pb
69

FLPIIAGIAAKVFPKIFCAISKKC




Brevinin-1Pc
70

FLPIIASVAAKVFSKIFCAISKKC




Brevinin-1Pd
71

FLPIIASVAANVFSKIFCAISKKC




Brevinin-1Pe
72

FLPIIASVAAKVFPKIFCAISKKC




Brevinin-1Pf
73

FLPIIAGIAAKFLPKIFCAISKKC




Brevinin-1Pk
74

FLPIIAGVAAKVFPKIFCTISKKC




Brevinin-1Pl
75

FLPIIAGMAAKFLPKIFCAISKKC




Brevinin-1SPb
76

FLPIIAGMAAKVICAITKKC




Brevinin-1Yb
77

FLPIIAGAAAKVVQKIFCAISKKC




Brevinin-1Yc
78

FLPIIAGAAAKVVEKIFCAISKKC






Temporins
Temporin-1AUa
79

FLPIIGQLLSGLL




Temporin-1BYa
80

FLPIIAKVLSGLL




Temporin-1DRc
81

FLPIIASVLSSLL




Temporin-1TGa
82

FLPIIGKLLSGIL




Temporin-PRb
83

FLPIITNLLGKLL






Vespid
Vespid 
84

FLPIIAKLVSGLL



Chemo-
chemotactic




tactic
peptide L




Peptides
Vespid 
85

FLPIIGKLLSGLL




chemotactic





peptide M





Vespid 
86

FLPIIAKLLGGLL




chemotactic





peptide X











In order to design a synthetic peptide with increased biological activity, the motif FLPII was engineered into the N-terminal extremity of mast-L (FIG. 1B), as changes in this part of the sequence are known to lead to loss of toxicity towards neutral vesicles mimicking human cell membranes(27) (Table 1).


Mastoparans, like other helical AMPs, are unstructured in aqueous solution and tend to structure into an a-helix upon contact with hydrophobic/hydrophilic interfaces, such as the one between bacterial membranes and the surrounding environment. The antimicrobial activity of such AMPs correlates with their ability to undergo the structural conformation change from unstructured to helical(7), and even small modifications to their sequence impact their structural tendency and biological activity. In order to assess whether insertion of the FLPII motif affected peptide structure, we elucidated the 3D structure of both mast-L and mast-MO through circular dichroism (CD— FIG. 2A) and of mast-MO using nuclear magnetic resonance (NMR— FIGS. 2B-2C and Table 2).









TABLE 2







Cytotoxic activity profile of mast-MO and mast-L. The cytotoxicity


of the peptides was evaluated against human red blood cells


(hRBCs), murine fibroblasts (L929), murine macrophages monocytes


(RAW 264.7), and human embryonic kidney (HEK293) cells.









Peptide Concentration (μmol L−1)












Cell Line
Mast- L
Mast-MO
LL-37
Gentamicin
Imipenem















hRBCs
7
>400
87
>400
>400


HEK293
50
25
NT
NT
NT


L929
10
>400
95
>400
>400


RAW 264.7
10
>400
100
>400
>400





NT—Not tested.






The CD experiments indicated that both peptides (FIG. 2) presented well-defined helical structures in the presence of TFE/Water (1:1; v:v), sodium dodecyl sulphate (SDS, 100 mmol L−1) micelles and phospholipid vesicles. The positive band around 192-193 nm and two negative bands at 208 and 222 nm (FIG. 2A) observed for both, mast-L and mast-MO, are characteristic profiles of a-helical structures(28). The highest a-helical content values, recorded for both peptides, was in SDS micelles (75.0% for mast-L and 79.5% for mast-MO), indicating that they undergo coil-helical transition upon contact with hydrophobic/hydrophilic interfaces, such as bacterial membranes. The same behavior was observed when peptides were added to other hydrophobic/hydrophilic interfaces such as dodecylphosphocholine (DPC) vesicles, trifluoroethanol (TFE)/Water mixture (3:2, v:v), and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC): 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG) (3:1, 10 mmol L−1) vesicle.


The a-helical structure of mast-MO was confirmed by 2D-NMR spectroscopy in deuterated SDS-d25 micelles (100 mmol L−1). The1I-1 resonance assignments were performed by simultaneous analyses of the TOCSY and NOESY contour maps (FIGS. 2B-2C), as previously described(29). The structural statistics for low-energy structures are detailed in Table 3.









TABLE 3







NMR and refinement statistics for peptide structures.










Mast-L
Mast-MO













NMR distance and dihedral constraints




Distance constraints


Total NOE
219
221


Intra-residue
135
101


Inter-residue


Sequential (|i − j| = 1)
72
113


Medium-range (|i − j| < 4)
12
7


Long-range (|i − j| > 5)
0
0


Intermolecular


Hydrogen bonds
0
0


Total dihedral angle restraints


ϕ
12
17


ψ
12
17


Structure statistics


Violations (mean and s.d.)
0.462 ± 0.011
0.525 ± 0.019


Distance constraints (Å)


Dihedral angle constraints (°)
0.343 ± 0.029
0.387 ± 0.028


Max. dihedral angle violation (°)
0.386
0.415


Max. distance constraint violation (Å)
0.483
0.557


Deviations from idealized geometry


Bond lengths (Å)
  0.018 ± 0.002 10−4
  0.018 ± 0.002 10−4


Bond angles (°)
0.462 ± 0.001
0.525 ± 0.003


Impropers (°)
0.343 ± 0.007
0.392 ± 0.005


Average pairwise RMS deviation** (Å)


Heavy
1.35 ± 0.31
1.42 ± 0.35


Backbone
0.76 ± 0.25
0.98 ± 0.30





**Pairwise RMS deviation was calculated among ten refined structures for residues 1 to 14 for mast-L and 1 to 19 for mast-MO.






The secondary structure of the peptides was predicted by chemical shift index (CSI) using NMRView, and the experimental chemical shifts of Ha, Ca, HN and N. CSI showed a well-defined a-helical structure throughout the whole sequence (between Leu3 and Leu14 residues) of mast-L and the maintenance of this helical behavior in mast-MO even with the additional of the pentapeptide motif (between Iles and Ile17 residues). The a-helical structure was confirmed using of the TALOS+software, in which the dihedral angles were estimated, phi


(4)) and psy (w) torsion angles (Table 4) of the peptide backbone and classified the values obtained as consistent with the predicted structures by CSI.









TABLE 4







Dihedral angle generated by the TALOS+.











Residue
Mast-L

Mast-MO












number
Phi (φ)
Psi (ψ)
Phi (φ)
Psi (ψ)














1






2
−81.987
−137.277 
−98.083
115.394


3
−61.350
−33.718
−60.487
145.120


4
−59.595
−42.256
−62.650
−24.572


5
−63.924
−41.425
−85.539
−16.800


6
−63.458
−40.117
−76.246
−25.646


7
−62.957
−36.890
−62.929
−40.922


8
−64.029
−41.696
−66.947
−35.670


9
−64.223
−42.331
−64.311
−44.022


10
−60.986
−42.934
−67.299
−39.006


11
−59.767
−35.453
−64.754
−42.410


12
−68.999
−36.486
−65.695
−36.129


13
−80.440
−24.618
−65.879
−39.182


14


−68.137
−35.730


15


−61.299
−41.414


16


−65.871
−34.946


17


−71.030
−35.612


18


−83.630
−16.631


19













Mastoparan peptides depend on their helical structure to exert antimicrobial activity(7). After initial electrostatic interactions with the membrane, this class of peptides tend to adopt an amphipathic helical structure with well-defined hydrophobic and hydrophilic faces upon contact with the phospholipids present on the membrane lipid bilayer, leading to membrane destabilization. Thus, the structural data showed that mast-MO preserved the helical tendency of mast-L, a critical requirement for these peptides to display antimicrobial activity.


Example 2— Assessment of Antimicrobial Activity, Immunomodulatory Effect, and Cytotoxicity


Synthetic peptide mast-MO exhibits decreased toxicity, and increased antimicrobial and immunomodulatory activities compared to parent wasp venom toxin mast-L. Helical conformations are known to be crucial for the antimicrobial activity of mastoparans(7). Once it was confirmed that insertion of the conserved pentapeptide motif FLPII did not alter the helical tendency of the peptide, a series of bacterial growth inhibition assays were performed in vitro to evaluate whether the motif influenced biological function. First, we assessed the minimal inhibitory concentration (MIC) of the peptide against 15 pathogens (FIG. 3A and FIG. 7). Pathogenic bacteria, including members of the ESKAPE list of the World Health Organization(2) and antibiotic-resistant strains, were exposed to increasing concentrations of the peptides for 24 h. Overall, mast-MO presented a similar activity profile, though slightly superior, compared to its predecessor mast-L, and increased activity with respect to two standard-of-care antibiotics (gentamicin and imipenem) (FIG. 3A). Briefly, mast-MO presented increased activity against Gram-positive bacteria compared to Gram-negatives and it was slightly more effective than mast-L against the majority of strains tested, with the exceptions of S. aureus MRSA, E. coli (2101123), E. coli 0157 and K pneumoniae 1825971 (KPC971). Mast-MO (4 μg mL−1) also exhibited increased antibiofilm properties against E. coli ATCC0157 and S. aureus ATCC33591 compared to mast-L both in vitro (FIG. 8) and in vivo (FIG. 6F).


Mastoparans are known to act through several mechanisms of action(13, 19-21, 30-32), and even minor modifications to their sequence have been shown to lead to different activity profiles(13, 31, 32). In order to gain insights into the mechanisms of action by which these peptides kill bacterial cells, we used the probes 3,3′-dipropylthiadicarbocyanine iodide [DiSC3(5)] and 1-(N-phenylamino)-naphthalene (NPN) to assess whether mast-MO was capable of depolarizing the cytoplasmic membrane and/or permeabilizing the outer membrane of bacteria, respectively. DiSC3(5) is a potentiometric fluorophore that accumulates in the cytoplasmic membrane of bacterial cells. When the accumulation of DiSC3(5) molecules reaches a certain threshold, those molecules aggregate, quenching their fluorescence. Upon disbalances on the transmembrane potential of the membrane, the fluorophore migrates to the cytoplasm or the outer environment, fluorescing again. In these assays, mast-MO only mildly depolarized the cytoplasmic membrane of 8 different clinically relevant Gram-negative bacterial strains (FIG. 3B), against which we had observed antimicrobial activity (FIG. 7). On the other hand, mast-MO potently permeabilized the outer membrane of the same set of 8 bacterial strains (FIG. 3C and FIG. 9) as determined by increased fluorescence of the lipophilic dye NPN in the NPN assay. NPN generates weak fluorescence in aqueous environments and is unable to permeate bacterial membranes unless they are damaged, but its fluorescence increases upon contact with lipidic environments such as the interior of the lipid bilayers of bacterial membranes therefore indicating that the membrane has been permeabilized and is thus compromised.


Once the mechanisms by which the peptide targeted bacterial cells was elucidawted, the effect of the pentapeptide motif insertion on cytotoxicity against mammalian cells was evaluated. Mast-MO did not present hemolytic activity in human red blood cell assays at the concentration range tested (1-400 μmoL−1), but it was toxic at 25μmoL−1 against the human embryonic cell line HEK293, whereas mast-L was both hemolytic and toxic towards the different human and m-urine cell lines tested at 7-50μmoL−1 (Table 2). Interestingly, mast-MO was not cytotoxic when incubated with mouse cell lines such as murine fibroblasts (L929 cells) and murine macrophages (RAW 264.7 cells) (Table 2). Therefore, the anti-infective activity of mast-MO was then tested in pre-clinical mouse models. First, the toxicity of a single dose of either mast-L or mast-MO administered peritoneally to mice was assessed in order to determine the range of concentrations to be used in subsequent animal experiments. The hemolytic activity of mast-MO (up to 200μmoL−1) against murine red blood cells (Table 6) was also tested prior to conducting animal experiments. In preliminary mouse experiments, mast-L was highly toxic at concentrations higher than 10 mg Kg−1 whereas mast-MO lacked toxicity (Table 5), in line with the presently disclosed results using mouse cell lines (Table 2).









TABLE 5







Evaluation of gross toxicity in mice treated with mastoparan-L


and mastoparan-MO. Each mouse was injected with the indicated


dose in 0.5 mL of peptide solution in PBS. Five animals


per group were directly inspected for adverse effects for


30 min, and mortality was monitored during 6 h.











Imipenem
Mast-L
Mast-MO



[number of
[number of
[number of


Dose (mg Kg−1)
mice (TL)]
mice (TL)]
mice (TL)]













0





1





5

2 (TL-1)



10

5



30

X



50
2 (TL-1)
X



90
3 (TL-2)
X
2 (TL-1)





— No observed effect, X all animals were dead, TL—Toxicity level: 1, narrowing of eyes; 2, crouching and cuddling. Most mice recovered 2 h after treatment.













TABLE 6







Evaluation of the hemolytic and cytotoxic activities of the motif


(FLPII) inserted in the N-terminal extremity of the synthetic peptide


mastoparan-MO. The hemolytic activity of mastoparan-MO against mouse


red blood cells (mRBCs) was assessed by exposing the cells to peptide


solutions ranging from 0.5 to 200 μmol L−1.


0.1% Triton X-100 was used as positive control (100% hemolysis)


and the human cathelicidin LL-37 was used for comparison.


The release of hemoglobin was measured at 550 nm and


expressed as percentage of lysed cells. The cytotoxic


activity of mastoparan-MO and LL-37 against RAW 264.7


monocytes was performed by incubating the cells for


24 h with peptide solutions ranging from 0.5 to 200


μmol L−1 and observing cell viability using MTT assays.


The data shown represent the mean of three biological replicates.










Peptide Concentration (μmol L−1)












Cells type
FLPII motif
LL-37







RAW 264.7
>200
100



mRBCs
>200
100










Based on these results, concentrations ranging from 1 to 10 mg Kg−1 of mast-MO and 1 to 5 mg Kg−1 of mast-L were used, which are not cytotoxic, in subsequent experiments.


To test whether the FLPII motif within the mast-MO sequence led to increased immunomodulatory and chemotactic properties with respect to mast-L, ELISA experiments were performed using the peritoneal fluid collected from mice (FIG. 5a). C57BL/6 mice were infected with both Gram-negative and Gram-positive pathogens and administered either mast-L or mast-MO intraperitoneally. For mice infected with E. coli ATCC8739, we monitored the peritoneal cavity to detect and quantify the release and presence of several immune mediators (FIG. 4E). Mast-MO-treated groups exhibited a pronounced ability to induce leukocyte migration to the site of infection, a key step into resolving infections, compared to the mast-L-treated group (FIG. 4B). This effect was similar to the positive control group of mice treated with transglutaminase (TGA) (FIG. 4B). Whereas mast-L did not significantly induce leukocyte recruitment over the time period tested, mast-MO stimulated recruitment at all time points with a peak of −108 observed 3 h post-infection (FIG. 4B). After 24 h, the leukocyte recruitment levels in mast-MO-treated mice dropped to −106, likely attributed to the resolution of the infection by the peptide (FIG. 5B and 5C). These results are consistent with the intrinsic ability of the pentapeptide motif FLPII to act as a chemoattractant (FIG. 1A), in line with our initial design, and highlight the immune boosting capability of our designed peptide. The concentration of interleukin-12 (IL-12), a protein produced mainly by macrophages and neutrophils in response to antigenic stimulation, and the pro-inflammatory cytokine tumor necrosis factor alpha (TNFa) were also longitudinally tracked for 48 h. Mast-MO and mast-L repressed both pro-inflammatory cytokines, in addition to IL-6, thus showing their ability to repress inflammation in vivo (FIG. 4C-4F). We monitored, via ELISAs, the cytokine profiles present within the peritoneal cavity of mice infected with E. coli ATCC8739 and S. aureus ATCC29213 (FIG. 4E and FIG. 4F, respectively) and treated with a single dose of peptide. Treatment with peptides in both cases led to a significant decrease in the inflammatory response within the peritoneal fluid resulting from the bacterial infection, thus highlighting the potential of the peptides as anti-inflammatory agents. These results confirmed that mast-MO appeared to control inflammation in addition to acting as a chemoattractant for leukocytes. Overall, these data suggest that mast-MO helps balance host responses by reducing the prolonged production of pro-inflammatory mediators while enhancing the activity and recruitment of leukocytes, which are important for baseline immune function.


Synthetic peptide mast-MO confers protection to mice against lethal septicemia caused by bacterial infections. To determine the anti-infective activity of mast-MO in a pre-clinical mouse model, the ability of the peptide to kill pathogenic and drug-resistant E. coli and S. aureus strains was tested in a systemic sepsis infection mouse model (FIG. 5). The minimal bacterial load resulting in a lethal dose (LDioo) 24 h post-intraperitoneal injection was first determined. The following LDioo were determined: −2x104 for E. coli ATCC8739 and E. coli 1812446 and 2×109 for S. aureus ATCC29213 and S. aureus (MRSA) ATCC33591. Mice were infected with the LDioo of bacteria and treated 3 h post-infection with either mast-L (1 and 5 mg Kg−1) or mast-MO (1, 5 and 10 mg Kg−1), which were also administered intraperitoneally. Mice infected with E. coli ATCC8739 and clinical isolate E. coli 1812446 (KPC-positive) and subsequently treated with mast-L (5 mg Kg−1) displayed decreased survival compared to groups treated with mast-MO (5 and 10 mg Kg−1) (FIG. 5B-5C and FIG. 5F-5G, respectively). Overall, mice treated with mast-MO exhibited 80% survival whereas those treated with mast-L displayed 40 to 60% survival. Mouse groups treated with mast-L at 10 mg Kg−1 had to be euthanized due to observed toxic effects when the peptide was injected systemically. Conversely, mice treated with mast-MO (10 mg Kg−1) were completely protected from infection by S. aureus ATCC29213 (FIG. 5D-5E), while those treated with mast-L (5 mg Kg−1) presented 80% survival. Finally, mice infected with S. aureus MRSA ATCC33591 and treated with mast-MO (10 mg Kg−1) exhibited 80% survival (FIG. 5H-5I). Only 60% of mice infected with S. aureus MRSA ATCC33591 and treated with mastoparan-L (5 mg Kg−1) survived. Once again, mice treated with 10 mg Kg−1 of mast-L had to be euthanized.


In summary, in all systemic infection experiments performed, 10 mg Kg′ of mast-MO led to significant reduction of bacterial counts and high survival rates, comparable to standard-of-care antibiotics, such as the aminoglycoside gentamicin and the (3-lactam imipenem, which are usually administered at 1-30 mg Kg′ in mouse models(33). These results further confirmed in vivo that mast-MO did not present toxicity in the mouse model tested, whereas the wasp venom peptide mast-L led to toxicity when injected systemically at 10 mg Kg′. Mast-MO, even at relatively low doses (10 mg Kgl), proved to be an excellent anti-infective agent, leading to complete inhibition of otherwise lethal bacterial infections caused by drug-resistant and clinically relevant Gram-negative and Gram-positive pathogens. The present in vivo data also demonstrated the dual activity of the presently disclosed engineered synthetic peptide mast-MO to both modulate the immune response and directly kill bacteria (FIG. 6A).


Example 3— Preparation and Assessment of Mast-MO Derivatives


In order to deplete the remaining toxicity observed in mast-MO towards certain human cells (e.g., HEK293; Table 2), the inventors designed and synthesized a permutation library of 59 novel mast-MO derivatives (Table 7) composed of single amino acid permutations of the FLPII motif (FIG. 6B).









TABLE 7







Amino acid sequences and aggregation in water of


the mast-MO analogs and other peptides from the


mastoparan family synthesized and tested in this


work.









Peptide
Sequence
Agg.





Galparan
GWTLNSAGYLLGPINL
NO


(SEQ ID NO: 2)
KALAALAKKIL






Transportan
GWTLNSAGYLLGKINL
NO


(SEQ ID NO: 3)
KALAALAKKIL






TP10 (SEQ ID NO: 4)
AGYLLGKINLKALAAL
NO



AKKIL



Mast-MO (SEQ ID
FLPIIINLKALAALAKKIL
NO


NO: 5)







 1 (SEQ ID NO: 6)
LFIIPINLKALAALAKKIL
NO





 2 (SEQ ID NO: 7)
IFLPIINLKALAALAKKIL
NO





 3 (SEQ ID NO: 8)
IIPFLINLKALAALAKKIL
NO





 4 (SEQ ID NO: 9)
LIPFIINLKALAALAKKIL
NO





 5 (SEQ ID NO: 10)
LIPIFINLKALAALAKKIL
NO





 6 (SEQ ID NO: 11)
LPIIFINLKALAALAKKIL
NO





 7 (SEQ ID NO: 12)
IIPLFINLKALAALAKKIL
NO





 8 (SEQ ID NO: 13)
ILIFPINLKALAALAKKIL
NO





 9 (SEQ ID NO: 14)
PLIFIINLKALAALAKKIL
++





10 (SEQ ID NO: 15)
PLIIFINLKALAALAKKIL
+





11 (SEQ ID NO: 16)
PILFIINLKALAALAKKIL
+





12 (SEQ ID NO: 17)
IPFILINLKALAALAKKIL
NO





13 (SEQ ID NO: 18)
IPLFIINLKALAALAKKIL
NO





14 (SEQ ID NO: 19)
LFIIPINLKALAALAKKIL
NO





15 (SEQ ID NO: 20)
IFLPIINLKALAALAKKIL
NO





16 (SEQ ID NO: 21)
IIPFLINLKALAALAKKIL
NO





17 (SEQ ID NO: 22)
LIPFIINLKALAALAKKIL
NO





18 (SEQ ID NO: 23)
ILFIPINLKALAALAKKIL
++





19 (SEQ ID NO: 24)
IFLIPINLKALAALAKKIL
NO





20 (SEQ ID NO: 25)
LIFIPINLKALAALAKKIL
NO





21 (SEQ ID NO: 26)
ILFPIINLKALAALAKKIL
NO





22 (SEQ ID NO: 27)
PFLIIINLKALAALAKKIL
+





23 (SEQ ID NO: 28)
FILIPINLKALAALAKKIL
NO





24 (SEQ ID NO: 29)
IFPLIINLKALAALAKKIL
NO





25 (SEQ ID NO: 30)
LIIPFINLKALAALAKKIL
NO





26 (SEQ ID NO: 31)
PIILFINLKALAALAKKIL
NO





27 (SEQ ID NO: 32)
FIPLIINLKALAALAKKIL
NO





28 (SEQ ID NO: 33)
IPFLIINLKALAALAKKIL
NO





29 (SEQ ID NO: 34)
IIFLPINLKALAALAKKIL
++





30 (SEQ ID NO: 35)
LIIFPINLKALAALAKKIL
NO





31 (SEQ ID NO: 36)
LFPIIINLKALAALAKKIL
NO





32 (SEQ ID NO: 37)
FLIPIINLKALAALAKKIL
NO





33 (SEQ ID NO: 38)
PFIILINLKALAALAKKIL
NO





34 (SEQ ID NO: 39)
IFIPLINLKALAALAKKIL
NO





35 (SEQ ID NO: 40)
FILPIINLKALAALAKKIL
NO





36 (SEQ ID NO: 41)
IILFPINLKALAALAKKIL
NO





37 (SEQ ID NO: 42)
PLFIIINLKALAALAKKIL
NO





38 (SEQ ID NO: 43)
FIPILINLKALAALAKKIL
NO





39 (SEQ ID NO: 44)
IFPILINLKALAALAKKIL
NO





40 (SEQ ID NO: 45)
IPILFINLKALAALAKKIL
NO





41 (SEQ ID NO: 46)
ILIPFINLKALAALAKKIL
+





42 (SEQ ID NO: 47)
IFILPINLKALAALAKKIL
NO





43 (SEQ ID NO: 48)
IPIFLINLKALAALAKKIL
NO





44 (SEQ ID NO: 49)
IILPFINLKALAALAKKIL
++





45 (SEQ ID NO: 50)
IPLIFINLKALAALAKKIL
NO





46 (SEQ ID NO: 51)
FIILPINLKALAALAKKIL
NO





47 (SEQ ID NO: 52)
FLIIPINLKALAALAKKIL
NO





48 (SEQ ID NO: 53)
PIFILINLKALAALAKKIL
+





49 (SEQ ID NO: 54)
FPIILINLKALAALAKKIL
NO





50 (SEQ ID NO: 55)
FIIPLINLKALAALAKKIL
NO





51 (SEQ ID NO: 56)
PIFLIINLKALAALAKKIL
NO





52 (SEQ ID NO: 57)
FPLIIINLKALAALAKKIL
NO





53 (SEQ ID NO: 58)
LPIFIINLKALAALAKKIL
++





54 (SEQ ID NO: 59)
PIIFLINLKALAALAKKIL
NO





55 (SEQ ID NO: 60)
LFIPIINLKALAALAKKIL
NO





56 (SEQ ID NO: 61)
ILPIFINLKALAALAKKIL
++





57 (SEQ ID NO: 62)
PILIFINLKALAALAKKIL
NO





58 (SEQ ID NO: 63)
FPILIINLKALAALAKKIL
NO





59 (SEQ ID NO: 64)
LIFPIINLKALAALAKKIL
++









The FLPII motif was chosen for the permutations to preserve the natural antimicrobial activity of mast-L and also conserve the hydrophobic content inserted in the N-terminal extremity for the initial design of mast-MO. Next, we assessed the antimicrobial (FIG. 10) and cytotoxic activities of the mast-MO analog library against HEK293 cells (FIG. 6C).


These specific cell line was chosen as it derives from human embryonic kidney cells and potential antimicrobial (i.e., AMPs, antibiotics) toxicity affects the kidneys, which are responsible for clearing such molecules (34). Also measured was whether the peptides aggregated at the highest concentration tested (128μmoL−1) (Table 7) since the motif inserted is composed of residues with aliphatic or aromatic side chain groups that may be prone to aggregation when in proximity within the primary peptide sequence, thus altering biological activity. Minor changes to the first five residues inserted in the mast-MO primary sequence led to a pronounced decrease in cytotoxic activity (2.5 to 5-fold) compared to mast-MO (FIG. 6C).


The most active peptides obtained, which were not cytotoxic and did not aggregate in aqueous solution, were selected and tested in a skin scarification mouse model that mimics an epithelial abscess infection(7, 12, 35). Mice were infected topically with the Gram-negative pathogen P. aeruginosa and subsequently treated with a single low dose (16 μmol L−1) of the seven lead synthetic peptides (analogs 14, 15, 26, 33, 47, 49, and 54— FIG. 6D-6F and Table 6). The peptides did not cause any side effects or adverse toxicity (i.e., hemolysis or cytotoxicity) towards mouse or human cell lines (FIG. 6E). Synthetic analogs (IFLPIINLKALAALAKKIL-NH2) (SEQ ID NO: 20) and 49 (FPIILINLKALAALAKKIL-NH2) (SEQ ID NO: 54) displayed increased anti-infective activity compared to their parent peptide mast-MO (FIG. 6E). Analog 15 is very similar to mast-MO as the permutation used to generate this molecule involved insertion an Ile residue at the beginning of the sequence while maintaining the last four residues of mast-MO. On the other hand, analog 49 presented all its aliphatic residues next to each other in positions three to five of the added motif, contrary to mast-MO that has a leucine residue in position two. Overall, several bactericidal mast-MO analogs were designed by permuting the 5 amino acid residues that compose the FLPII and 2 of these analogs presented high anti-infective activity with a single dosage in a skin abscess formation mouse model at low micromolar concentration (16 μmol L−1), comparable to the most active anti-infective peptides(7, 12).


Materials and Methods


Peptide synthesis. Mast-L, mast-MO and LL-37 were purchased from Shanghai Hanhong Chemical (China) and Mast-MO analogs were purchased from Biopolymers (MIT). The peptides were synthesized using the solid-phase peptide synthesis and N-9-fluoromethyloxycarbonyl (Fmoc) strategy and purified by high-performance liquid chromatography (HPLC). Peptide purity used in biologic assays was higher than 95%.


Circular Dichroism spectroscopy. The circular dichroism (CD) experiments were performed to determine the ideal conditions for carrying out the NMR experiments, including conformational preferences and the stability ratio at different pH and media concentrations. Mast-L at 70μmoL−1 and mast-MO at 50μmoL−1 were analyzed in SDS (100 mmol L−1), DPC (15 mmol L−1), TFE/Water (1:1, v:v), and PCPG (1 mmol L−1) phospholipid vesicles, at 20° C. The spectra were recorded on a Jasco J-815 spectropolarimeter coupled to a Peltier Jasco PTC-423L (Tokyo, Japan), using a 1.0 mm path length rectangular quartz cuvette (NSG, Farmingdale N.Y.), six accumulations from 260 to 190 nm, 1.0 nm spectral bandwidth, 0.2 nm step resolution, 100 nm min−1 scan speed and 1 s response time. Similar experiments with the respective blank solutions were performed for background subtraction. The spectra were analyzed using the CDPro software package (36, 37). The relative helix content (fp) was calculated from the ellipticity values at 222 nm as described by Chen et al.(38)


NMR spectroscopy and structure calculations. The samples were prepared by dissolving the peptide stock solution (1 mmol L−1) in 500 μL H20/D20 (9/1, v/v) in a micellar solution containing 100 mmol L−1 of sodium dodecyl sulfate (SDS-d25), pH 4.0 and 25° C. All spectra were recorded on a Bruker Avance III 500 spectrometer equipped with a 5 mm broadband inverse (BBI) probehead. Proton chemical shifts were referenced to trimethylsilyl-2,2,3,3-d4-propionate sodium (TMSP-d4).



1H-1H TOCSY experiments were recorded with 40 transients of 4096 data points, 512 tl increments and spinlock mixing time of 80 ms. 1H-1H NOESY spectra were recorded with 64 transients of 1024 data points, 512 tl increments and mixing time of 100, 150, 200 and 250 ms. 1H-13C HSQC experiments, edited mode, were acquired with spectral widths Fl of 20831 and F2 of 8012 Hz, and 80 transients of 4096 points for each free induction decay and 256 tl increments were used. 1H-15N sf-HMQC (Heteronuclear multiple-quantum coherence) experiments were recorded with 1024 data points and 80 tl increments, 7,600 transients for mastoparan-L and 7,000 for mastoparan-MO. All 2D-NMR data were processed using NMRPIPE(39) and analyzed with NMRView(40) software.


The 1H-1H NOESY spectra revealed a total of 219 distance restraints for mast-L and 221 for mast-MO, with 15.6 and 11.6 average restrictions per residue, respectively. The ten structures obtained for each of the mastoparan peptides were highly convergent, i.e. mast-MO is a well-structured helical peptide. We also observed that when each one of the peptides had their structured region aligned, they presented RMSD values of 0.47±0.15 A and 0.41±0.18 A, for mast-L and mast-MO respectively. Despite the random character of the N-terminal region, the RMSD of all aligned amino acid residues was 0.76±0.25 and 0.98±0.30 A (mean±standard deviation of nine samples), for mast-L and mast-MO, respectively. This indicates a geometric distribution and conformational flexibility of both mastoparans. The average of the ten Ramachandran plots for mast-L shows that 94.2% of the cp and w angles are in most favored a-helical regions and 5.8% in allowed regions, whereas mast-MO exhibits 100% of the 4) and tlf angles in the most favored a-helical regions (http://mordred.bioc.cam.ac.uk/—rapper/rampage.php). These results indicated that the calculated structures were predicted accurately. Additionally, we checked the fold accuracy using ProSA II(41), which indicated Z-score values of -6.00 and -3.56 for mast-MO and mast-L, respectively. These values indicated the absence of erroneous parts of our models, which points to the accuracy of the structure prediction.


H20/D20 exchange kinetics experiments were performed using a solution of 1 mM of the peptide in the presence of 100 mmol L−1 of SDS-d25 diluted in H20/D20 (1:9, v/v) to a final volume of 600 pt. TMSP-d4 was used as the reference standard. Thirty minutes were spent between the preparation of the sample and the beginning of the data collection. TOCSY experiments were performed every 2 h at the temperature of 298 K for 7 day.


The structure calculations and refinement were performed using the program XPLOR-NIH version 2.28 by simulated annealing (SA) algorithm(42). NOE cross peak volumes were converted into semi-quantitative distance restrains using the calibration by Hyberts et al.(43). The upper limits of the distances restrain obtained were calibrated for 2.8, 3.4, and 5.0 A, being strong, medium, and weak NOE, respectively. Once attributed the NOE correlations and the chemical shifts of the backbone atoms in the respective contour maps, these were respectively converted into distance and dihedral angle restrains, which were employed in simulated annealing calculations to generate the structures of the mastoparan peptides. Both phi (c)) and psi (w) backbone dihedral angles were predicted, by analysis of 1Ha and 13Ca, using TALOS+(44). Starting with an extended conformation, 200 structures were calculated, 20 structures were selected, from the 200 calculated structures, and refined in water (42). The 10 lowest energy conformations were selected.


Protein Structure Validation Suite (PSVS 5.2 server) was used to evaluate the quality of the structures (http://psys-1_5 -dev.nesg.org/). The stereochemical quality of the lowest energy structures, was evaluated using PROCHECK through the Ramachandran plot (http://mordred.bioc.cam.ac.uk/—rapper/rampage.php) and ProSA (Protein Structure Analysis) indicated the fold quality Z-scores. The display, analysis, manipulation of the molecular graphics images and root mean square deviation (RMSD) calculations were performed using either PyMOL (The PyMOL Molecular Graphics System, http://www.pymol.org) and MOLMOL(45) software.


Bacterial strains and media. Strains used included clinical isolates Escherichia coli KPC-positive ID No . 1812446, Escherichia coli multidrug-resistant ID 2101123 and carbapenemase-producing Klebsiella pneumoniae KPC-positive ID 1825971, as well as reference strains Bacillus subtilis ATCC6633, Enterococcus faecalis ATCC12953, Staphylococcus aureus ATCC29213, Methicillin-Resistant Staphylococcus aureus ATCC33591, Streptococcus pyogenes ATCC19615, Escherichia coli ATCC8739, Klebsiella pneumoniae ATCC13885, Proteus mirabilis ATCC25933, Pseudomonas aeruginosa ATCC 15442, Salmonella enterica ATCC14028, Acinetobacter baumannii ATCC19606, Escherichia coli ATCC11775, Escherichia coli AIG221, Escherichia coli AIG222, Escherichia coli BL21, Klebsiella pneumoniae ATCC133883, Pseudomonas aeruginosa PAO1, Pseudomonas aeruginosa PA14 and Salmonella enterica RFP. Bacteria were plated on brain heart infusion broth (BHI) (Himedia, India) from a frozen stock. Following 24 h of incubation of the agar plate, three isolated colonies were transferred to 1 mL of BHI. The broth culture was incubated overnight (12-16 h) at 37° C. with shaking.


Antibacterial assays. Minimum inhibitory concentration (MIC) of peptides and antibiotics were evaluated using the broth microdilution technique in Mueller-Hinton Broth medium (MHB) with an initial inoculum of 5×105 cells in nontreated Polystyrene microtiter plates (Corning, USA) in accordance with Wiegand et al. The MIC was interpreted as the lowest concentration of peptide or antibiotic that completely inhibited the visible growth of bacteria after 12 h of incubation of the plates at 37° C. Each agent was tested in triplicate in at least three independent experiments. The MIC assays were also performed using the broth microdilution method in sterile 96-well polypropylene microtiter plates. Peptides were added to the plate as solutions in BM2 minimal medium in concentrations ranging from 0 to 128 μg.mL−1, and the bacteria (E. coli BL21, S. aureus ATCC12600, P. aeruginosa PA01 and PA14) were grown overnight at 37° C. and inoculated at a final concentration of 5×105 CFU mL-1 per well. The plates were incubated at 37° C. for 24 h and read in a plate reader at 610 nm. All assays were done in triplicate.


Bacterial killing assays. The bacterial killing experiments involved performing 1:100 dilutions of overnight cultures of E. coli BL21, S. aureus ATCC12600, P. aeruginosa PAO1 and PA14 in the absence or presence of increasing concentrations of the samples (0-200 μg mL−1). After 24 h of treatment, 10-fold serial dilutions were performed, bacteria were plated on LB agar plates (E. coli BL21 and S. aureus ATCC12600) and Pseudomonas Isolation Agar (P. aeruginosa PAO1 and PA14) and allowed to grow overnight at 37° C. after which colony forming unit (CFU) counts were recorded. All experiments were done in triplicates.


Antibiofilm assays. Experiments were performed as described previously. Biofilms were grown in BM2 glucose medium for 72 h, at 37° C. in flow cell chambers with channel dimensions of 1×4×40 mm, as previously described by de la Fuente-Nunez et al.. For the treatment of pre-formed biofilms, bacteria were allowed to develop structured 2-day-old biofilms prior to treatment with peptides for the following 24 h. Biofilm cells were then stained using the Live/Dead BacLight bacterial viability kit (Molecular Probes, Eugene, Oreg.) and subsequently examined using a confocal laser scanning microscope (Olympus, Fluoview FV1000); three-dimensional reconstructions were generated using the Imaris software package (Bitplane AG).


Hemolysis assays. Hemolytic activity of peptides was determined by using fresh mouse red blood cells (mRBCs), by measuring the peptide-induced changes of the optical density (OD) at 540 nm (Victor X, Perkin-Elmer, Germany), 100% lysis was determined by analysing the supernatant of erythrocytes that had been incubated with Triton X-100 (1%).


Mammalian cells toxicity assays. L929 mice fibroblasts (Rio de Janeiro Cell Bank) and SPCs were seeded in 96-well microtiter plates in a concentration of 1.0×105 cells per well, in DMEM medium, supplemented with different concentrations of tested peptides (1-600 04). After 48 h incubation, a thiazolyl blue tetrazolium bromide (MTT) protocol was performed. Briefly, 60% of the medium was removed, and 10 μL of MTT (5 mg mL−1) (Sigma, USA) solution was added to each well and the plate was incubated for 4 h, in 5% CO2, at 37° C. The blue formazan product generated was dissolved by the addition of 100 μL of 100% DMSO (Mallinckrodt, Germany) per well. Plates were then gently swirled for 5 min, at room temperature, to dissolve the precipitate. The absorbance was monitored at 575 nm using a microplate spectrophotometer (Bio-Tek, USA). Cytotoxicity was determined as a percentage of the maximum value after subtracting the background. The results were expressed as the percentage of each sample compared to the negative control (PBS buffer, pH 7.4) and cell culture was incubated in a lysis buffer (10 mM Tris, pH 7.4, 1 mM ethylenediamine tetraacetic acid (EDTA), and 0.1% Triton X-100).


Membrane permeabilization assays. The membrane permeability of the mast-MO (20 mol L−1) was determined by using the N-phenyl-1-napthylamine (NPN) uptake assay. All the bacterial strains were grown to OD600 of 0.4, centrifuged (10,000 rpm at 4° C. for 10 min), washed and resuspended in buffer (5 mmol L−1 HEPES, 5 mmol L−1 glucose, pH 7.4). 4 μL of NPN solution (0.5 mmol L−1— working concentration of 10 wino′ L−1 after dilutions) was added to 100 μL of the bacterial solution in a white 96-wells plate. The background fluorescence was recorded at Xex=350 nm and Xem=420 nm. Mast-MO samples in water (100 μL solution at 1.2 mol L−1) were added to the 96-wells plate, and fluorescence was recorded as a function of time until no further increase in fluorescence was observed (20 min).


Membrane depolarization assays. The cytoplasmic membrane depolarization activity of mast-MO (20 mol L−1) was determined by the membrane potential—sensitive dye, DiSC3(5). Briefly, the bacterial strains were grown at 37° C. with agitation to the mid-log phase (OD600=0.5). The cells were centrifuged and washed twice with washing buffer (20 mmol L−1 glucose, 5 mmol L−1 HEPES, pH 7.2) and re-suspended to an OD600 of 0.05 in the same buffer (20 mmol L−1 glucose, 5 mmol L−1 HEPES, pH 7.2) but containing 0.1 mol L−1 KCl. Thereafter, the cells (100 μL) were incubated for 15 min with 20 nmol L−1 of DiSC3(5) until a stable reduction of fluorescence was achieved, indicating the incorporation of the dye into the bacterial membrane. Membrane depolarization was then monitored by observing the change in the fluorescence emission intensity of the membrane potential—sensitive dye, DiSC3(5) (Xex=622 nm, Xem=670 nm).


Bacterial toxicity mouse model. Six-weeks-old female C57BL/6 mice were used in the murine systemic infection model. Animals were provided by the Central


Bioterium of the Universidade de Sao Paulo - Ribeirao Preto. All animals were housed in individual cages under a constant temperature (22° C.) and humidity with a 12 h light/dark cycle and had access to food and water ad libitum throughout the study. The mice were euthanized by CO2 at the end of the experiments. All procedures, care, and handling of the animals were approved by the Ethics Committee of the Catholic University of Brasilia number 005/13. CD-1 IGS female mice (6-weeks-old) were used for the skin scarification model and maintained in accordance with the Guide for the Care and Use of Laboratory Animals in an AAALAC-accredited facility.


Systemic bacterial infection mouse model. Acute toxicity assay was performed based on the work of Navon-Venezia and co-workers(46). The experiment was performed by intraperitoneal (i.p.) injection of the tested peptides to groups of 10 C57BL/6 mice. Each mouse was injected with a 0.5 mL solution of freshly prepared of peptides in PBS. The doses of peptide administered per mouse were 0, 10, 30, 50, 70 and 90 mg Kg′ of body weight. Animals were directly inspected for adverse effects for 6 h, and mortality was monitored for 7 days thereafter. Differences between groups were analysed using the Fisher exact test (differences were considered to be statistically significant when the P value was <0.05). Briefly, E. coli ATCC8739 and E. coli 1812446 or S. aureus ATCC 29213 and S. aureus (MRSA) ATCC33591 were plated on tryptic soy agar plus 5% sheep blood (blood agar, Himedia, India) from a frozen stock. Following 24 h of incubation of the agar plate, three isolated colonies were transferred to 1 mL of BHI. The broth culture was incubated overnight (12-16 h) at 37° C. with shaking. Based on preliminary experiments to determine the challenge dose of E. coli ATCC8739 and E. coli KPC-positive ID 1812446 or S. aureus ATCC29213 and S. aureus (MRSA) ATCC33591 that resulted in consistent systemic infection without rapidly killing the mice, the broth-grown bacteria were diluted to −2×104 (CFU/mL per mouse in phosphate buffered saline— PBS) for Gram-negative bacteria and 2×109(CFU/mL per mouse in PBS) for Gram-positive bacteria. Mice were challenged with either strain of E. coli ATCC8739 and E. coli 1812446 or S. aureus ATC29213 and S. aureus (MRSA) ATCC33591 in a final volume of 200 μL of PBS by intraperitoneal injection (i.p.). The day of challenge was designated as day 1 of the experiment. Three hours after bacterial injection, mice (n=5) were treated, intraperitoneally, with different concentrations of peptides (1, 5 or 10 mg Kg1), LL-37, gentamicin and 10 mg Kg′ of imipenem or PBS for 8 days. For bacterial load evaluation, 5 mice per group were used. Animals were humanely killed 24 h after bacterial administration. To detect bacterial colonization, the peritoneal lavage samples were collected under sterile conditions. The peritoneal lavage was obtained by washing the cavity with 5 mL of sterile PBS. Samples were diluted serially, and 100 μL of each dilution was spread in duplicate on appropriate agar plates for the count of developed colonies. Cytokine levels (i.e., IFN-γ, IL-6, IL-10, IL-12p70 and TNF-α) in the peritoneal lavage of the mice both in the absence and presence of peptide treatment were quantified by ELISA (Peprotech, USA) according to the manufacturer's instructions.


Isolation of leukocytes from the peritoneal cavity of mice. Leukocyte migration into the peritoneal cavity of uninfected C57BL/6 mice treated with 10 mg Kg′ of peptides was evaluated to assess the chemotactic activity of the peptides, as previously described(11). Specifically, 10 mg Kg−1 of peptides suspended in sterile saline were injected i.p. in C57BL/6 mice. Mice were subsequently euthanized, and their peritoneal lavage was collected over time. Leukocyte counts present in the in peritoneal cavity of mice infected with E. coli ATCC 8739 were quantified. Animals infected and treated with peptides were euthanized over time (0.5-24 h) post-treatment, and a peritoneal lavage was carried out to quantify leukocyte numbers present in the peritoneal cavity(47). Thioglycolate (TGA) 3% was used as a positive control for leukocyte migration. Animals were sacrificed, and cells present in the peritoneal cavity were harvested in 3 mL of PBS containing 1 mmol L-1 EDTA. Total cell counts were quantified using a cell counter (Coulter AC T series analyzer), and differential cell counts were conducted on cytocentrifuge slides (Cytospin 3; Thermo Shandon) stained by the May-Grunwald-Giemsa (Rosenfeld) method. The results are expressed as the number of cells per cavity(48).


Skin abscess infection mouse model. P. aeruginosa strain PAO1 was used for inducing a skin infection in the mouse model as described by Pane et al.(9). Briefly, bacteria were grown in tryptic soy broth medium. Subsequently, cells were washed twice with sterile PBS (pH 7.4, 13,000 rpm for 1 min), and resuspended to a final concentration of 1×107 CFU/20 μL. Female CD-1 mice (six-weeks old) were anesthetized with isoflurane and had their backs shaved and a superficial linear skin abrasion was made with a needle in order to damage the stratum corneum and upper-layer of the epidermis. An aliquot of 20 containing 1×107 CFU of bacteria in PBS was inoculated over each defined area containing the scratch with a pipette tip. One day after the infection, peptides (16 mol L−1) were administered to the infected area. Animals were euthanized and the area of scarified skin was excised two- and four-days post-infection, homogenized using a bead beater for 20 minutes (25 Hz), and serially diluted for CFU quantification. Two independent experiments were performed with 3 mice per group in each condition (n=6 per group).


REFERENCES

The following publications may also be relevant to the present disclosure:

  • 1. de la Fuente-Nunez C, Torres MD, Mojica FJ, Lu TK (2017) Next-generation precision antimicrobials: towards personalized treatment of infectious diseases. Curr Opin Microbiol 37:95-102.
  • 2. Centers for Disease Control and Prevention (2019) 2019 Antimicrobial Resistant


Threats Report Available at: https://www.cdc.gov/drugresistance/biggest-threats.html.

  • 3. Rudd KE, et al. (2020) Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet 395(10219):200-211.
  • 4. Morens DM, Taubenberger JK, Fauci AS (2008) Predominant Role of Bacterial


Pneumonia as a Cause of Death in Pandemic Influenza: Implications for Pandemic Influenza Preparedness. J Infect Dis 198(7):962-970.

  • 5. Zhou F, et al. (2020) Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 395(10229):1054-1062.
  • 6. Lewis RJ, Garcia ML (2003) Therapeutic potential of venom peptides. Nat Rev Drug


Discov 2(10):790-802. 7 Torres MDT, et al. (2018) Structure-function-guided exploration of the antimicrobial peptide polybia-CP identifies activity determinants and generates synthetic therapeutic candidates. Commun Biol 1:221.

  • 8. Torres MDT, Sothiselvam S, Lu TK, de la Fuente-Nunez C (2019) Peptide Design


Principles for Antimicrobial Applications. J Mol Biol:In Press.

  • 9. Cardoso MH, et al. (2018) A Computationally Designed Peptide Derived from Escherichia coli as a Potential Drug Template for Antibacterial and Antibiofilm Therapies. ACS Infect Dis 4(12):1727-1736.
  • 10. Candido ES, et al. (2019) Short Cationic Peptide Derived from Archaea with Dual


Antibacterial Properties and Anti-Infective Potential. ACS Infect Dis doi:10.1021/acsinfecdis.9b00073.

  • 11. Silva ON, et al. (2016) An anti-infective synthetic peptide with dual antimicrobial and immunomodulatory activities. Sci Rep 6(1):35465.
  • 12. Porto WF, et al. (2018) In silico optimization of a guava antimicrobial peptide enables combinatorial exploration for peptide design. Nat Commun 9(1):1490.
  • 13. Oshiro KGN, et al. (2019) Computer-Aided Design of Mastoparan-like Peptides


Enables the Generation of Nontoxic Variants with Extended Antibacterial Properties. J Med Chem 62(17):8140-8151.

  • 14. Torres MDT, et al. (2017) Decoralin Analogs with Increased Resistance to Degradation and Lower Hemolytic Activity. ChemistrySelect 2(1):18-23.
  • 15. Nijnik A, Hancock R (2009) Host defence peptides: antimicrobial and immunomodulatory activity and potential applications for tackling antibiotic-resistant infections. Emerg Health Threats J 2. doi:10.3134/ehtj 0.09.001.
  • 16. Lima SMF, et al. (2017) Antimicrobial and immunomodulatory activity of host defense peptides, clavanins and LL-37, in vitro: An endodontic perspective. Peptides 95:16-24.
  • 17. Kim WH, Lillehoj HS, Min W (2017) Evaluation of the Immunomodulatory Activity of the Chicken NK-Lysin-Derived Peptide cNK-2. Sci Rep 7(1):45099.
  • 18. Lazar V, et al. (2018) Antibiotic-resistant bacteria show widespread collateral sensitivity to antimicrobial peptides. Nat Microbiol 3(6):718-731.
  • 19. Higashijima T, Uzu S, Nakajima T, Ross EM (1988) Mastoparan, a peptide toxin from wasp venom, mimics receptors by activating GTP-binding regulatory proteins (G proteins). J Biol Chem 263(14):6491-4.
  • 20. Mousli M, et al. (1989) Activation of rat peritoneal mast cells by substance P and mastoparan. J Pharmacol Exp Ther 250(1):329-35.
  • 21. Higashijima T, Burnier J, Ross EM (1990) Regulation of Gi and Go by mastoparan, related amphiphilic peptides, and hydrophobic amines. Mechanism and structural determinants of activity. J Biol Chem 265(24):14176-86.
  • 22. Wang G, Li X, Wang Z (2016) APD3: the antimicrobial peptide database as a tool for research and education. Nucleic Acids Res 44(D1):D1087—D1093.
  • 23. Conlon J M, et al. (2009) Peptides with potent cytolytic activity from the skin secretions of the North American leopard frogs, Lithobates blairi and Lithobates yavapaiensis. Toxicon 53(7-8):699-705.
  • 24. Goraya J, et al. (2000) Peptides with antimicrobial activity from four different families isolated from the skins of the North American frogs Rana luteiventris, Rana berlandieri and Rana pipiens. Eur J Biochem 267(3):894-900.
  • 25. Yang X, Wang Y, Lee W-H, Zhang Y (2013) Antimicrobial peptides from the venom gland of the social wasp Vespa tropica. Toxicon 74:151-157.
  • 26. Hernandez-Gras F, Boronat A (2015) A hydrophobic proline-rich motif is involved in the intracellular targeting of temperature-induced lipocalin. Plant Mol Biol 88(3):301-311.
  • 27. Barany-Wallje E, Gaur J, Lundberg P, Lange′ U, Graslund A (2007) Differential membrane perturbation caused by the cell penetrating peptide Tpl 0 depending on attached cargo. FEBS Lett 581(13):2389-2393.
  • 28. Greenfield NJ (2006) Using circular dichroism spectra to estimate protein secondary structure. Nat Protoc 1(6):2876-2890.
  • 29. Wiithrich K (1986) NMR of Proteins and Nucleic Acids ed Sons JW (New York).
  • 30. Irazazabal LN, et al. (2016) Selective amino acid substitution reduces cytotoxicity of the antimicrobial peptide mastoparan. Biochim Biophys Acta - Biomembr 1858(11):2699-2708.
  • 31. Souza BM de, et al. (2015) Structure—activity relationship of mastoparan analogs:


Effects of the number and positioning of Lys residues on secondary structure, interaction with membrane-mimetic systems and biological activity. Peptides 72:164-174.

  • 32. Chen X, et al. (2018) Evaluation of the bioactivity of a mastoparan peptide from wasp venom and of its analogues designed through targeted engineering. Int.1 Biol Sci 14(6):599-607.
  • 33. Pletzer D, Mansour SC, Wuerth K, Rahanj am N, Hancock REW (2017) New Mouse


Model for Chronic Infections by Gram-Negative Bacteria Enabling the Study of Anti-Infective Efficacy and Host-Microbe Interactions. MBio 8(1):e00140-17.

  • 34. Brouwer CPJM, Wulferink M, Welling MM (2008) The Pharmacology of


Radiolabeled Cationic Antimicrobial Peptides. J Pharm Sci 97(5):1633-1651.

  • 35. Pane K, et al. (2018) Identification of Novel Cryptic Multifunctional Antimicrobial


Peptides from the Human Stomach Enabled by a Computational—Experimental Platform. ACS Synth Biol 7(9):2105-2115.

  • 36. Sreerama N, Woody RW (2000) Estimation of protein secondary structure from circular dichroism spectra: Comparison of CONTIN, SELCON, and CDSSTR methods with an expanded reference set. Anal Biochem 287(2):252-260.
  • 37. Woody RW (2004) On the analysis of membrane protein circular dichroism spectra.


Protein Sci 13:100-112.

  • 38. Chen YH, Yang JT, Chau KH (1974) Determination of the helix and beta form of proteins in aqueous solution by circular dichroism. Biochemistry 13(16):3350-9.
  • 39. Delaglio F, et al. (1995) NMRPipe: a multidimensional spectral processing system based on UNIX pipes. J Biomol NMR 6(3):277-93.
  • 40. Johnson BA, Blevins RA (1994) NMR View: A computer program for the visualization and analysis of NMR data. J Biomol NMR 4(5):603-14.
  • 41. Wiederstein M, Sippl MJ (2007) ProSA-web: interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic Acids Res 35:407-410.
  • 42. Schwieters CD, Kuszewski JJ, Tjandra N, Marius Clore G (2003) The Xplor-NIH


NMR molecular structure determination package. J Magn Reson 160(1):65-73.

  • 43. Hyberts SG, Goldberg MS, Havel TF, Wagner G (1992) The solution structure of eglin c based on measurements of many NOEs and coupling constants and its comparison with X-ray structures. Protein Sci 1(6):736-51.
  • 44. Shen Y, Delaglio F, Comilescu G, Bax A (2009) TALOS+: a hybrid method for predicting protein backbone torsion angles from NMR chemical shifts. J Biomol NMR 44(4):213-23.
  • 45. Koradi R, Billeter M, Wilthrich K (1996) MOLMOL: a program for display and analysis of macromolecular structures. J Mol Graph 14(1):51-5,29-32.
  • 46. Navon-Venezia S, Feder R, Gaidukov L, Carmeli Y, Mor A (2002) Antibacterial


Properties of Dermaseptin S4 Derivatives with In Vivo Activity. Antimicrob Agents Chemother 46(3):689-694.

  • 47. Ray A, Dittel BN (2010) Isolation of Mouse Peritoneal Cavity Cells. J Vis Exp (35):e1488.
  • 48. Moreno SE, et al. (2006) IL-12, but Not IL-18, Is Critical to Neutrophil Activation and Resistance to Polymicrobial Sepsis Induced by Cecal Ligation and Puncture. J Immunol 177(5):3218-3224.
  • 49. Vert M, et al. (2012) Terminology for biorelated polymers and applications (IUPAC Recommendations 2012). Pure Appl Chem 84(2):377-410.
  • 50. de la Fuente-Nffiez C, Reffuveille F, Fernandez L, Hancock REW (2013) Bacterial biofilm development as a multicellular adaptation: antibiotic resistance and new therapeutic strategies. Curr Opin Microbiol 16(5):580-589.
  • 51. Boase NRB, Torres MDT, Fletcher NL, de la Fuente-Nunez C, Fairfull-Smith KE (2018) Polynitroxide copolymers to reduce biofilm fouling on surfaces. Polym Chem. doi:10.1039/C8PY01101J.
  • 52. Pizzo E, et al. (2018) Novel bioactive peptides from PD-L1/2, a type 1 ribosome inactivating protein from Phytolacca dioica L. Evaluation of their antimicrobial properties and anti-biofilm activities. Biochim Biophys Acta - Biomembr 1860(7). doi:10.1016/j.bbamem.2018.04.010.

Claims
  • 1. An antimicrobial peptide that comprises a mastoparan peptide having SEQ ID NO:1 and a pentapeptide motif formed from phenylalanine, leucine, proline, and two isoleucine residues, wherein the pentapeptide motif is conjugated the N-terminus of the mastoparan peptide.
  • 2. The antimicrobial peptide according to claim 1 comprising any one of SEQ ID NOS:5-64.
  • 3. The antimicrobial peptide according to claim 1 comprising any one of SEQ ID NOS:6-64.
  • 4. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 19.
  • 5. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 20.
  • 6. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 31.
  • 7. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 38.
  • 8. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 52.
  • 9. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 54.
  • 10. The antimicrobial peptide according to claim 1 comprising SEQ ID NO: 59.
  • 11. A method of treating a microbial infection in a subject comprising administering to a subject a therapeutically effective amount of an antimicrobial peptide according to claim 1.
  • 12. A method comprising contacting a biofilm with an effective amount of an antimicrobial peptide according to claim 1.
  • 13. A method of forming an antimicrobial peptide comprising conjugating a pentapeptide motif formed from phenylalanine, leucine, proline, and two isoleucine residues to a mastoparan peptide having SEQ ID NO:1 at the N-terminus of the peptide.
  • 14. The method according to claim 13, wherein the antimicrobial peptide comprises any one of SEQ ID NOS:5-64.
  • 15. The method according to claim 13 comprising any one of SEQ ID NOS:6-64.
  • 16. The method according to claim 13 comprising SEQ ID NO: 19.
  • 17. The method according to claim 13 comprising SEQ ID NO: 20.
  • 18. The method according to claim 13 comprising SEQ ID NO: 31.
  • 19. The method according to claim 13 comprising SEQ ID NO: 38.
  • 20. The method according to claim 13 comprising SEQ ID NO: 52.
  • 21. The method to claim 13 comprising SEQ ID NO: 54.
  • 22. The method according according to claim 13 comprising SEQ ID NO: 59.
  • 23. A composition comprising an antimicrobial peptide according to claim 1 and a pharmaceutically acceptable carrier.
  • 24. The composition according to claim 23, comprising two or more antimicrobial peptides comprising SEQ ID NOS:5-64.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims benefit to U.S. Provisional Application No. 63/243,845, filed Sep. 14, 2021, the entire contents of which are incorporated herein by reference.

Provisional Applications (1)
Number Date Country
63243845 Sep 2021 US