Throughout this disclosure, various patent and technical publications are identified by an identifying citation or an Arabic numeral, the full citations for which are found immediately preceding the claims. These citations and the publications referenced within the present specification are incorporated by reference into the present disclosure to more fully describe the state of the art to which this invention pertains.
Innate immunity is the first line of host defense. In response to invading pathogens, pattern recognition receptors (PRRs) sense pathogen-associated molecular patterns (PAMPs) that are structural components or replication intermediates (Medzhitov, 2007; Takeuchi and Akira, 2010; Ting et al., 2010). PRRs include the cytosolic receptors (e.g., cGAS, IFI16, RIG-I-like and NOD-like receptors) and the membrane-anchored Toll-like receptors (TLRs) and C-type lectins. Upon binding to PAMPs, PRRs recruit cognate adaptor molecules that signal to activate two closely-related kinase complexes, IKKα/β and TBK-1/IKKε. IKKα/β phosphorylates and induces the degradation of the inhibitor of NF-κB (IκBs), leading to the nuclear translocation of NF-κB (Chen et al., 1996; Zandi et al., 1997). TBK-1/IKKε can directly phosphorylate interferon regulatory factors (IRFs) to induce its dimerization and translocation into the nucleus (Fitzgerald et al., 2003; Sharma et al., 2003). Along with other transcription factors, nuclear NF-κB and IRFs coordinate to up-regulate the expression of many immune genes to engender an antiviral state (Bhatt and Ghosh, 2014). The cytosolic RIG-I receptor is a genuine RNA sensor that, in response to viral infection, activates NF-κB and IRFs through the mitochondrion antiviral signaling (MAVS) protein (Kawai et al., 2005; Meylan et al., 2005; Seth et al., 2005; Xu et al., 2005). Studies entailing gene knockout mice demonstrate that loss of RIG-I or MAVS severely impairs host innate immune response and greatly increases viral replication (Kato et al., 2006; Sun et al., 2006). Not surprisingly, viruses have evolved diverse strategies to halt or hijack antiviral signaling downstream of RIG-I and MAVS (Chan and Gack, 2015; Feng et al., 2013).
Post-translational modification (PTM) is a major means to regulate protein function and underpins diverse fundamental biological processes. First reported more than five decades ago (Mycek and Waelsch, 1960), deamidation of asparagine/glutamine in protein has long been regarded as a non-specific process associated with protein “aging”. Early protein deamidation research surveyed the overall deamidation of the cellular proteome, and led to the postulate that non-enzymatic protein deamidation serves as a biological clock for protein “aging” (Robinson and Robinson, 2001; Weintraub and Deverman, 2007). As such, research in protein deamidation is scarce and accordingly Applicant's understanding is rudimentary at best. A few proteins (e.g., Bcl-xL and 4EBP2) were shown to be regulated by deamidation in mammalian cells, which was postulated to be the consequence of an increase in cellular pH (Bidinosti et al., 2010; Deverman et al., 2002; Dho et al., 2013). Recent studies demonstrate that pathogenic bacteria secrete effectors to deamidate key signaling molecules to evade host immune defenses (Cui et al., 2010; Sanada et al., 2012) and manipulate cellular signaling (Flatau et al., 1997; Schmidt et al., 1997), indicating that protein deamidation can be catalyzed by bacterial enzymes and is highly regulated. The roles of protein deamidation in metazoan remain largely unclear.
It has been reported that gamma herpesviruses, including human Kaposi's sarcoma-associated herpesvirus (KSHV) and murine gamma herpesvirus 68 (γHV68), deploy vGAT pseudo-enzymes to induce RIG-I deamidation (He et al., 2015; Kolakofsky and Garcin, 2015). Though lacking intrinsic enzyme activity, vGAT proteins recruited cellular phosphoribosylformyglycinamidine synthetase (PFAS, also known as FGARAT) to deamidate and concomitantly activate RIG-I. Activated RIG-I was harnessed by γHV68 to evade antiviral cytokine production (Dong and Feng, 2011; Dong et al., 2012). Applicant reports here that herpes simplex virus 1 (HSV-1) induces RIG-I deamidation to prevent RIG-I activation by viral dsRNA. The UL37 tegument protein was sufficient to deamidate RIG-I in cells and in vitro, making it the first viral protein deamidase to be identified. Site-specific deamidation within the helicase 2i domain impaired the RNA detection and ATP hydrolysis of RIG-I. Uncoupling RIG-I deamidation from HSV-1 infection restored RIG-I activation and anti-viral cytokine production, thereby reducing HSV-1 replication. This work delineates a pivotal role of protein deamidation in sensing nucleic acid by a PRR and demonstrates that HSV-1 exploits protein deamidation to evade innate immune defense.
The therapeutic and prophylactic interventions and screens disclosed herein are derived from Applicant's disclosed discoveries. As background, RIG-I detects double-stranded RNA (dsRNA) to trigger antiviral cytokine production. Protein deamidation is emerging as a post-translational modification that chiefly regulates protein function. Applicant reports here that UL37 of herpes simplex virus 1 (HSV-1) is a protein deamidase that targets RIG-I to block RNA-induced activation. Mass spectrometry analysis identified two asparagine residues in the helicase 2i domain that were deamidated upon UL37 expression or HSV-1 infection. Deamidations in the helicase 2i domain rendered RIG-I unable to sense viral dsRNA, trigger antiviral immune responses and restrict viral replication. Purified full-length UL37 and its carboxyl terminal fragment were sufficient to deamidate RIG-I in vitro. Uncoupling RIG-I deamidation from HSV-1 infection, via engineering deamidation-resistant RIG-I or introducing deamidase-deficient UL37 into the HSV-1 genome, restored RIG-I activation and antiviral immune signaling. This work defines the first viral deamidase and a pivotal role of protein deamidation in sensing microbial pathogens by a pattern recognition receptor.
This disclosure provides an isolated polynucleotide encoding a RIG-I mutant and equivalents thereof as well as compliment thereto. In one aspect, the RIG-I mutant is RIG-I-QQ. A non-limiting example of this polypeptide is SEQ ID NO. 4, and equivalents thereof and complements thereto. An equivalent is one or more polypeptide that retains amino acids at positions 495 and/or 549 that make the protein deaminase resistant, e.g., a substitution of Q at positions 495 and/or 549. Vectors and host cells are further provided herein. Complementary polypeptides to these are further provided herein.
Further provided are polypeptides are encoded by polynucleotides.
Compositions containing one or more of the polynucleotides, proteins, vectors and host cells and one or more carriers, are further provided herein. In one aspect, the compositions contain buffers, stabilizers and/or preservatives. In a further aspect, the compositions are lyophilized for ease of transport, storage and use.
The compositions can be formulated for administration as a vaccine or therapeutic composition and contain an effective immunity-inducing amount of the active components and optionally an adjuvant. The compositions can be further formulated into dosage units that can be packaged into kits with instructions for use.
The compositions are useful in methods to inhibit viral replication by contacting the virus in a cell, tissue or subject in need thereof by with an effective amount of an agent that inhibits the deamidation activity of UL37. Also provided is a method to abolish 5′-ppp-RNA-binding and ATP hydrolysis is a cell, tissue or subject infected with the virus, as well as to inhibit or “switch off RIG-1 by contacting the cell, tissue or administering to the subject an effective amount of a composition as provided herein. In another aspect, provided herein is a method to block RNA-induced activation by a cell, tissue or subject in need thereof by contacting the cell, tissue or administering to the subject. In a further aspect, this disclosure also provides a method to induce an anti-viral immune response in a subject in need thereof by administering an effective amount of a composition as described herein.
In one aspect, the virus is an virus that exhibits these activities such as a DNA virus, e.g., a virus of the class Herpesviridae, e.g., HIV-1 or HSV-2 virus.
Methods to determine if the methods are effective are known in the art and disclosed herein, e.g., a reduction in viral load, deamination assay, ATPase activation assay, enhanced immunity, e.g., B-cell or T-cell adaptive immunity, etc.
The contacting can be in vitro or in vivo, and the administration can be effected by methods known in the art, e.g., injection or oral administration. Multiple administrations can be provided as necessary.
For in vivo methods, the subject to be treated is any subject at risk of or having a viral infection, e.g., a pet, sports animal or human patient.
Attached are nucleotide sequences that are relevant to disclosure;
SEQ ID NO.: 1 is the wild-type polynucleotide sequence of UL37.
SEQ ID NO.: 2 is the mutated polynucleotide sequence designated UL37 C819S.
SEQ ID NO.: 3 depicts wild-type RIGI polypeptide.
SEQ ID NO.: 4 depicts mutated RIG-I-QQ polypeptide.
SEQ ID NO.: 5 depicts the polynucleotide sequence of Strain KOS of HSV-1, a mutated HSV-1 having mutated UL37.
Before the compositions and methods are described, it is to be understood that the invention is not limited to the particular methodologies, protocols, cell lines, assays, and reagents described, as these may vary. It is also to be understood that the terminology used herein is intended to describe particular embodiments of the present invention, and is in no way intended to limit the scope of the present invention as set forth in the appended claims.
Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods, devices, and materials are now described. All technical and patent publications cited herein are incorporated herein by reference in their entirety. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of tissue culture, immunology, molecular biology, microbiology, cell biology and recombinant DNA, which are within the skill of the art. See, e.g., Sambrook and Russell eds. (2001) Molecular Cloning: A Laboratory Manual, 3rd edition; the series Ausubel et al. eds. (2007) Current Protocols in Molecular Biology; the series Methods in Enzymology (Academic Press, Inc., N.Y.); MacPherson et al. (1991) PCR 1: A Practical Approach (IRL Press at Oxford University Press); MacPherson et al. (1995) PCR 2: A Practical Approach; Harlow and Lane eds. (1999) Antibodies, A Laboratory Manual; Freshney (2005) Culture of Animal Cells: A Manual of Basic Technique, 5th edition; Gait ed. (1984) Oligonucleotide Synthesis; U.S. Pat. No. 4,683,195; Hames and Higgins eds. (1984) Nucleic Acid Hybridization; Anderson (1999) Nucleic Acid Hybridization; Hames and Higgins eds. (1984) Transcription and Translation; Immobilized Cells and Enzymes (IRL Press (1986); Perbal (1984) A Practical Guide to Molecular Cloning; Miller and Calos eds. (1987) Gene Transfer Vectors for Mammalian Cells (Cold Spring Harbor Laboratory); Makrides ed. (2003) Gene Transfer and Expression in Mammalian Cells; and Mayer and Walker eds. (1987) Immunochemical Methods in Cell and Molecular Biology (Academic Press, London).
All numerical designations, e.g., pH, temperature, time, concentration, and molecular weight, including ranges, are approximations which are varied (+) or (−) by increments of 0.1. It is to be understood, although not always explicitly stated that all numerical designations are preceded by the term “about”. It also is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.
As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above.
As used in the specification and claims, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a cell” includes a plurality of cells, including mixtures thereof.
As used herein, the term “comprising” or “comprises” is intended to mean that the compositions and methods include the recited elements, but not excluding others. “Consisting essentially of” when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives and the like. “Consisting of” shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention or process steps to produce a composition or achieve an intended result. Embodiments defined by each of these transition terms are within the scope of this invention.
The term “isolated” as used herein with respect to nucleic acids, such as DNA or RNA, refers to molecules separated from other DNAs or RNAs, respectively that are present in the natural source of the macromolecule. The term “isolated peptide fragment” is meant to include peptide fragments which are not naturally occurring as fragments and would not be found in the natural state. The term “isolated” is also used herein to refer to polypeptides, antibodies, proteins, host cells and polynucleotides that are isolated from other cellular proteins or tissues and is meant to encompass both purified and recombinant polypeptides, antibodies, proteins and polynucleotides. In other embodiments, the term “isolated” means separated from constituents, cellular and otherwise, in which the cell, tissue, polynucleotide, peptide, polypeptide, protein, antibody or fragment(s) thereof, which are normally associated in nature and can include at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95%, or alternatively at least 98%, purified from a cell or cellular extract. For example, an isolated polynucleotide is separated from the 3′ and 5′ contiguous nucleotides with which it is normally associated in its native or natural environment, e.g., on the chromosome. An isolated cell, for example, is a cell that is separated form tissue or cells of dissimilar phenotype or genotype. As is apparent to those of skill in the art, a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody or fragment(s) thereof, does not require “isolation” to distinguish it from its naturally occurring counterpart.
The term “binding” or “binds” as used herein are meant to include interactions between molecules that may be detected using, for example, a hybridization assay. The terms are also meant to include “binding” interactions between molecules. Interactions may be, for example, protein-protein, antibody-protein, protein-nucleic acid, protein-small molecule or small molecule-nucleic acid in nature. This binding can result in the formation of a “complex” comprising the interacting molecules. A “complex” refers to the binding of two or more molecules held together by covalent or non-covalent bonds, interactions or forces.
Hybridization reactions can be performed under conditions of different “stringency”. In general, a low stringency hybridization reaction is carried out at about 40° C. in about 10×SSC or a solution of equivalent ionic strength/temperature. A moderate stringency hybridization is typically performed at about 50° C. in about 6×SSC, and a high stringency hybridization reaction is generally performed at about 60° C. in about 1×SSC. Hybridization reactions can also be performed under “physiological conditions” which is well known to one of skill in the art. A non-limiting example of a physiological condition is the temperature, ionic strength, pH and concentration of Mg2+ normally found in a cell.
The term “polypeptide” is used interchangeably with the term “protein” and in its broadest sense refers to a compound of two or more subunit amino acids, amino acid analogs or peptidomimetics. The subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g., ester, ether, etc. As used herein the term “amino acid” refers to natural and/or unnatural or synthetic amino acids, including glycine and both the D and L optical isomers, amino acid analogs and peptidomimetics. A peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein. The term “peptide fragment” as used herein, also refers to a peptide chain.
The phrase “equivalent polypeptide” or “biologically equivalent peptide or peptide fragment” or “biologically equivalent polynucleotide” refers to a protein or a peptide fragment which is homologous to the exemplified reference polynucleotide, protein or peptide fragment and which exhibit similar biological activity in vitro or in vivo, e.g., approximately 100%, or alternatively, over 90% or alternatively over 85% or alternatively over 70%, as compared to the standard or control biological activity. Additional embodiments within the scope of this invention are identified by having more than 60%, or alternatively, more than 65%, or alternatively, more than 70%, or alternatively, more than 75%, or alternatively, more than 80%, or alternatively, more than 85%, or alternatively, more than 90%, or alternatively, more than 95%, or alternatively more than 97%, or alternatively, more than 98% or 99% sequence identity or homology. Percentage homology can be determined by sequence comparison using programs such as BLAST run under appropriate conditions. In one aspect, the program is run under default parameters.
The term “polynucleotide” refers to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides or analogs thereof. Polynucleotides can have any three-dimensional structure and may perform any function, known or unknown. The following are non-limiting examples of polynucleotides: a gene or gene fragment (for example, a probe, primer, or EST), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, RNAi, siRNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes and primers. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polynucleotide. The sequence of nucleotides can be interrupted by non-nucleotide components. A polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component. The term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form.
A polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for thymine when the polynucleotide is RNA. Thus, the term “polynucleotide sequence” is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching.
“Homology” or “identity” or “similarity” are synonymously and refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An “unrelated” or “non-homologous” sequence shares less than 40% identity, or alternatively less than 25% identity, with one of the sequences of the present invention.
A polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) has a certain percentage (for example, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99%) of “sequence identity” to another sequence means that, when aligned, that percentage of bases (or amino acids) are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in Ausubel et al. eds. (2007) Current Protocols in Molecular Biology. Preferably, default parameters are used for alignment. One alignment program is BLAST, using default parameters. In particular, programs are BLASTN and BLASTP, using the following default parameters: Genetic code=standard; filter=none; strand=both; cutoff=60; expect=10; Matrix=BLOSUM62; Descriptions=50 sequences; sort by=HIGH SCORE; Databases=non-redundant, GenBank+EMBL+DDBJ+PDB+GenBank CDS translations+SwissProtein+SPupdate+PIR. Details of these programs can be found at the following Internet address: http://www.ncbi.nlm.nih.gov/blast/Blast.cgi, last accessed on Nov. 26, 2007. Biologically equivalent polynucleotides are those having the specified percent homology and encoding a polypeptide having the same or similar biological activity.
The term “non-contiguous” refers to the presence of an intervening peptide, nucleotide, polypeptide or polynucleotide between a specified region and/or sequence. For example, two polypeptide sequences are non-contiguous because the two sequences are separated by a polypeptide sequences that is not homologous to either of the two sequences. Non-limiting intervening sequences are comprised of at least a single amino acid or nucleotide.
A “gene” refers to a polynucleotide containing at least one open reading frame (ORF) that is capable of encoding a particular polypeptide or protein after being transcribed and translated. Any of the polynucleotide or polypeptide sequences described herein may be used to identify larger fragments or full-length coding sequences of the gene with which they are associated. Methods of isolating larger fragment sequences are known to those of skill in the art.
The term “express” refers to the production of a gene product such as RNA or a polypeptide or protein.
As used herein, “expression” refers to the process by which polynucleotides are transcribed into mRNA and/or the process by which the transcribed mRNA is subsequently being translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in an eukaryotic cell.
The term “encode” as it is applied to polynucleotides refers to a polynucleotide which is said to “encode” a polypeptide if, in its native state or when manipulated by methods well known to those skilled in the art, it can be transcribed and/or translated to produce the mRNA for the polypeptide and/or a fragment thereof. The antisense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced there from.
Applicant have provided herein the polypeptide and/or polynucleotide sequences for use in gene and protein transfer and expression techniques described below. It should be understood, although not always explicitly stated that the sequences provided herein can be used to provide the expression product as well as substantially identical sequences that produce a protein that has the same biological properties. These “biologically equivalent” or “biologically active” polypeptides are encoded by equivalent polynucleotides as described herein. They may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide when compared using sequence identity methods run under default conditions. Specific polypeptide sequences are provided as examples of particular embodiments. Modifications to the sequences to amino acids with alternate amino acids that have similar charge.
A polynucleotide of this invention can be delivered to a cell or tissue using a gene delivery vehicle. “Gene delivery,” “gene transfer,” “transducing,” and the like as used herein, are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a “transgene”) into a host cell, irrespective of the method used for the introduction. Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection/transfection, or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of “naked” polynucleotides (such as electroporation, “gene gun” delivery and various other techniques used for the introduction of polynucleotides). The introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome. A number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
A “composition” is intended to mean a combination of active polypeptide, polynucleotide or antibody and another compound or composition, inert (e.g. a detectable label) or active (e.g. a gene delivery vehicle) alone or in combination with a carrier which can in one embodiment be a simple carrier like saline or pharmaceutically acceptable or a solid support as defined below.
A “pharmaceutical composition” is intended to include the combination of an active polypeptide, polynucleotide or antibody with a carrier, inert or active such as a solid support, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
As used herein, the term “pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Martin (1975) Remington's Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton).
A “subject,” “individual” or “patient” is used interchangeably herein, and refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, rats, rabbits, simians, bovines, ovines, porcines, canines, felines, farm animals, sport animals, pets, equines, and primates, particularly humans.
“Cell,” “host cell” or “recombinant host cell” are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. The cells can be of any one or more of the type murine, rat, rabbit, simian, bovine, ovine, porcine, canine, feline, equine, and primate, particularly human.
Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
Herpesviridae is a large family of DNA viruses that cause diseases in animals, including humans. Non-limiting examples of the members include HSV-1, HSV-2, varicella zoster virus, Epstein-Barr virus, cytomegalovirus, varicella-zoster virus, human herpesvirus 6A and 6B, and Karposi's sarcoma-associated herpesvirus.
“Treating,” “treatment,” or “ameliorating” of a disease includes: (1) preventing the disease, i.e., causing the clinical symptoms of the disease not to develop in a patient that may be predisposed to the disease but does not yet experience or display symptoms of the disease; and/or (2) inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; and/or (3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
The term “suffering” as it related to the term “treatment” refers to a patient or individual who has been diagnosed with or is predisposed to a disease or infection by a virus. A patient may also be referred to being “at risk of suffering” from a disease or infection by a virus. This patient has not yet developed characteristic disease pathology, however are known to be predisposed to the disease due to family history, being genetically predispose to developing the disease, or diagnosed with a disease or disorder that predisposes them to developing the disease to be treated.
An “effective amount” is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages. Such delivery is dependent on a number of variables including the time period for which the individual dosage unit is to be used, the bioavailability of the therapeutic agent, the route of administration, etc. It is understood, however, that specific dose levels of the therapeutic agents disclosed herein for any particular subject depends upon a variety of factors including the activity of the specific compound employed, bioavailability of the compound, the route of administration, the age of the animal and its body weight, general health, sex, the diet of the animal, the time of administration, the rate of excretion, the drug combination, and the severity of the particular disorder being treated and form of administration. In general, one will desire to administer an amount of the compound that is effective to achieve a serum level commensurate with the concentrations found to be effective in vivo. These considerations, as well as effective formulations and administration procedures are well known in the art and are described in standard textbooks.
“Under transcriptional control” is a term well understood in the art and indicates that transcription of a polynucleotide sequence, usually a DNA sequence, depends on its being operatively linked to an element which contributes to the initiation of, or promotes, transcription. “Operatively linked” intends the polynucleotides are arranged in a manner that allows them to function in a cell.
A “probe” when used in the context of polynucleotide manipulation refers to an oligonucleotide that is provided as a reagent to detect a target potentially present in a sample of interest by hybridizing with the target. Usually, a probe will comprise a detectable label or a means by which a label can be attached, either before or subsequent to the hybridization reaction. Alternatively, a “probe” can be a biological compound such as a polypeptide, antibody, or fragments thereof that is capable of binding to the target potentially present in a sample of interest.
“Detectable labels” or “markers” include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes. Detectable labels can also be attached to a polynucleotide, polypeptide, antibody or composition described herein.
A “primer” is a short polynucleotide, generally with a free 3′—OH group that binds to a target or “template” potentially present in a sample of interest by hybridizing with the target, and thereafter promoting polymerization of a polynucleotide complementary to the target. A “polymerase chain reaction” (“PCR”) is a reaction in which replicate copies are made of a target polynucleotide using a “pair of primers” or a “set of primers” consisting of an “upstream” and a “downstream” primer, and a catalyst of polymerization, such as a DNA polymerase, and typically a thermally-stable polymerase enzyme. Methods for PCR are well known in the art, and taught, for example in MacPherson et al. (1991) PCR 1: A Practical Approach (IRL Press at Oxford University Press). All processes of producing replicate copies of a polynucleotide, such as PCR or gene cloning, are collectively referred to herein as “replication.” A primer can also be used as a probe in hybridization reactions, such as Southern or Northern blot analyses. Sambrook and Russell (2001), infra.
“Hybridization” refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. A hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
Hybridization reactions can be performed under conditions of different “stringency”. In general, a low stringency hybridization reaction is carried out at about 40° C. in 10×SSC or a solution of equivalent ionic strength/temperature. A moderate stringency hybridization is typically performed at about 50° C. in 6×SSC, and a high stringency hybridization reaction is generally performed at about 60° C. in 1×SSC. Hybridization reactions can also be performed under “physiological conditions” which is well known to one of skill in the art. A non-limiting example of a physiological condition is the temperature, ionic strength, pH and concentration of Mg2 normally found in a cell.
When hybridization occurs in an antiparallel configuration between two single-stranded polynucleotides, the reaction is called “annealing” and those polynucleotides are described as “complementary”. A double-stranded polynucleotide can be “complementary” or “homologous” to another polynucleotide, if hybridization can occur between one of the strands of the first polynucleotide and the second. “Complementarity” or “homology” (the degree that one polynucleotide is complementary with another) is quantifiable in terms of the proportion of bases in opposing strands that are expected to form hydrogen bonding with each other, according to generally accepted base-pairing rules.
The term “propagate” means to grow a cell or population of cells. The term “growing” also refers to the proliferation of cells in the presence of supporting media, nutrients, growth factors, support cells, or any chemical or biological compound necessary for obtaining the desired number of cells or cell type.
The term “culturing” refers to the in vitro propagation of cells or organisms on or in media of various kinds. It is understood that the descendants of a cell grown in culture may not be completely identical (i.e., morphologically, genetically, or phenotypically) to the parent cell.
A “viral vector” is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro. Examples of viral vectors include retroviral vectors, lentiviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like. Alphavirus vectors, such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy. See, Schlesinger and Dubensky (1999) Curr. Opin. Biotechnol. 5:434-439 and Ying, et al. (1999) Nat. Med. 5(7):823-827.
In aspects where gene transfer is mediated by a lentiviral vector, a vector construct refers to the polynucleotide comprising the lentiviral genome or part thereof, and a therapeutic gene. As used herein, “lentiviral mediated gene transfer” or “lentiviral transduction” carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome. The virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell. Retroviruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell. The integrated DNA form is called a provirus. As used herein, lentiviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism. A “lentiviral vector” is a type of retroviral vector well-known in the art that has certain advantages in transducing nondividing cells as compared to other retroviral vectors. See, Trono D. (2002) Lentiviral vectors, New York: Spring-Verlag Berlin Heidelberg.
Lentiviral vectors of this invention are based on or derived from oncoretroviruses (the sub-group of retroviruses containing MLV), and lentiviruses (the sub-group of retroviruses containing HIV). Examples include ASLV, SNV and RSV all of which have been split into packaging and vector components for lentiviral vector particle production systems. The lentiviral vector particle according to the invention may be based on a genetically or otherwise (e.g. by specific choice of packaging cell system) altered version of a particular retrovirus.
That the vector particle according to the invention is “based on” a particular retrovirus means that the vector is derived from that particular retrovirus. The genome of the vector particle comprises components from that retrovirus as a backbone. The vector particle contains essential vector components compatible with the RNA genome, including reverse transcription and integration systems. Usually these will include gag and pol proteins derived from the particular retrovirus. Thus, the majority of the structural components of the vector particle will normally be derived from that retrovirus, although they may have been altered genetically or otherwise so as to provide desired useful properties. However, certain structural components and in particular the env proteins, may originate from a different virus. The vector host range and cell types infected or transduced can be altered by using different env genes in the vector particle production system to give the vector particle a different specificity.
“RNA interference” (RNAi) refers to sequence-specific or gene specific suppression of gene expression (protein synthesis) that is mediated by short interfering RNA (siRNA).
“Short interfering RNA” (siRNA) refers to double-stranded RNA molecules (dsRNA), generally, from about 10 to about 30 nucleotides in length that are capable of mediating RNA interference (RNAi), or 11 nucleotides in length, 12 nucleotides in length, 13 nucleotides in length, 14 nucleotides in length, 15 nucleotides in length, 16 nucleotides in length, 17 nucleotides in length, 18 nucleotides in length, 19 nucleotides in length, 20 nucleotides in length, 21 nucleotides in length, 22 nucleotides in length, 23 nucleotides in length, 24 nucleotides in length, 25 nucleotides in length, 26 nucleotides in length, 27 nucleotides in length, 28 nucleotides in length, or 29 nucleotides in length. As used herein, the term siRNA includes short hairpin RNAs (shRNAs).
“Double stranded RNA” (dsRNA) refer to double stranded RNA molecules that may be of any length and may be cleaved intracellularly into smaller RNA molecules, such as siRNA. In cells that have a competent interferon response, longer dsRNA, such as those longer than about 30 base pair in length, may trigger the interferon response. In other cells that do not have a competent interferon response, dsRNA may be used to trigger specific RNAi.
The term siRNA includes short hairpin RNAs (shRNAs). shRNAs comprise a single strand of RNA that forms a stem-loop structure, where the stem consists of the complementary sense and antisense strands that comprise a double-stranded siRNA, and the loop is a linker of varying size. The stem structure of shRNAs generally is from about 10 to about 30 nucleotides in length. For example, the stem can be 10-30 nucleotides in length, or alternatively, 12-28 nucleotides in length, or alternatively, 15-25 nucleotides in length, or alternatively, 19-23 nucleotides in length, or alternatively, 21-23 nucleotides in length.
Tools to assist siRNA design are readily available to the public. For example, a computer-based siRNA design tool is available on the internet at www.dharmacon.com, Ambion-www.ambion.com/jp/techlib/misc/siRNA_finder.html; Thermo Scientific-Dharmacon-www.dharmacon.com/DesignCenter/DesignCenterPage.aspx; Bioinformatics Research Center-sysbio.kribb.re.kr:8080/AsiDesigner/menuDesigner.jsf; and Invitrogen-maidesigner.invitrogen.com/maiexpress/.
As used herein, the term “purification label” refers to at least one marker useful for purification or identification. A non-exhaustive list of this marker includes His, lacZ, GST, maltose-binding protein, NusA, BCCP, c-myc, CaM, FLAG, GFP, YFP, cherry, thioredoxin, poly(NANP), V5, Snap, HA, chitin-binding protein, Softag 1, Softag 3, Strep, or S-protein. Suitable direct or indirect fluorescence marker comprise FLAG, GFP, YFP, RFP, dTomato, cherry, Cy3, Cy 5, Cy 5.5, Cy 7, DNP, AMCA, Biotin, Digoxigenin, Tamra, Texas Red, rhodamine, Alexa fluors, FITC, TRITC or any other fluorescent dye or hapten.
Applicant has identified a mechanism by which certain virus evade a host's innate immune response. Provided herein are compositions and methods that build upon this discovery. To that end, in one aspect provided herein is an isolated polynucleotide encoding a RIG-I-QQ mutant and equivalents thereof. Non-limiting examples of equivalents include polynucleotides that hybridize under stringen conditions to the polynucleotide (e.g., a polynucleotide encoding SEQ ID NO. 4) and sequences having at least 70% sequence identity to a polynucleotide encoding SEQ ID NO. 4). In one aspect, the isolated polynucleotide encodes the polypeptide shown in SEQ ID NO. 4, and equivalents that retain amino acids at positions 495 and/or 549 that make the protein deaminase resistant, e.g., substitution of Q at position positions 495 and/or 549. The isolated polynucleotides can be included within a vector or other gene delivery vehicle or isolated host cell. The RIG-I-QQ polypeptide and equivalents thereof can be combined or contained with a host cell and/or with a carrier, such as a pharmaceutically acceptable carrier. The polypeptide and proteins can be chemically and/or recombinantly produced using methods known in the art, using host cells containing the polynucleotides and culturing the cell under conditions for expression and/or replication of the polynucleotides or polypeptides. The polypeptides and polynucleotides can be further combined with a detectable label or a purification label and used for purification and/or in drug development screens. Compositions containing the RIG-I-QQ mutant or an equivalent thereof can be combined with other antiviral agents and immune enhancing compositions such as an vaccine adjuvant.
Also provided herein is an isolated mutated UL37 polynucleotide that fails to deaminate RIG-I polypeptide. Non-limiting examples of such is the polynucleotide is a modified wild-type UL 37 mutated at positions 819 and 850, e.g., identified herein as UL 37 C819S (SEQ ID NO.: 2) and C850S, and equivalents thereof. The isolated mutated UL37 polynucleotides fail to deaminate RIG-I polypeptide. The mutated polynucleotides can be combined with a label, e.g., a detectable or purification label for screening, probes, primers or other assays. The polynucleotides can be chemically or recombinantly produced using methods known in the art. In one aspect, they are combined within an HSV vector or virus and are useful in vaccine compositions. In one aspect, they are combined with a carrier, such as a pharmaceutically acceptable carrier and/or adjuvant. The compositions can be incorporated into a kit and can further contain instructions for use, e.g., in the methods disclosed herein.
The agents and compositions of the present disclosure can be used in the manufacture of medicaments and for the treatment of humans and other animals by administration in accordance with conventional procedures, such as an active ingredient in pharmaceutical compositions.
Also provided are methods for one or more of:
The contacting can be performed in vitro or in vivo. When performed in vivo, the agent is administered to a subject infected with the virus or for whom prophylaxis is desired. Any suitable method of administration can be used in the method, e.g., topical, intravenous, by inhalation therapy. The subject is any animal that is susceptible to the viral infection e.g., a mammal or a human. The method can further comprise administration of an effective amount of an antiviral agent.
Administration can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the infection being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. Route of administration can also be determined and method of determining the most effective route of administration are known to those of skill in the art and will vary with the composition used for treatment, the purpose of the treatment, the health condition or disease stage of the subject being treated, and target cell or tissue. Non-limiting examples of route of administration include oral administration, nasal administration, injection, topical application, intraperitoneal, intravenous and by inhalation.
Also provided herein is a method for one or more of:
“Administration” can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the virus being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. Route of administration can also be determined and method of determining the most effective route of administration are known to those of skill in the art and will vary with the composition used for treatment, the purpose of the treatment, the health condition or disease stage of the subject being treated, and target cell or tissue. Non-limiting examples of route of administration include oral administration, nasal administration, injection, topical application, intraperitoneal, intravenous and by inhalation.
HEK293T, HeLa, Vero, HCT116, BHK21, mouse embryonic fibroblasts (MEFs) and human foreskin fibroblasts (HFF) were cultured in Dulbecco's modified Eagle's medium (DMEM, Corning) supplemented with 10% heat-inactivated fetal bovine serum (FBS; HyClone), penicillin (100 U/mL) and streptomycin (100 μg/mL). Wild-type and Rig-i−/− MEFs were described previously (Kato et al., 2005). Wild-type HSV-1 (KOS strain), GFP HSV-1 and HSV-1 recombinant viruses were amplified in Vero cells, with viral titers ranging from 107 to 108 pfu/ml. eGFP VSV (Dr. Sean Whelan) was amplified in BHK21 cells, with viral titer of 109 pfu/ml. Sendai virus was purchased from Charles River Laboratories.
Luciferase reporter plasmids for the NF-κB, IFN-β promoter, PRDIII (ISRE) promoter, mammalian expression plasmids for RIG-I and their truncated mutants, MDA5, MAVS, IKKβ, TBK1, IRF3-5D, RelA were described previously (Dong et al., 2010; Dong and Feng, 2011; Dong et al., 2012; He et al., 2015; Seth et al., 2005). The non-silencing (control) shRNA and shRNA against human RIG-I, human IFI16 and human STING were purchased from Thermo Scientific. HSV-1 expression library was described previously (Sen et al., 2013). Mammalian expression plasmids for truncated RIG-I and UL37, lentiviral expression plasmids for RIG-I and UL37 were generated by standard molecular biology techniques. All point mutants, including those of RIG-I and UL37, were generated by site-directed mutagenesis and confirm by sequencing. HSV-1ΔUL37 (KOS) and HSV-1(KOS) Bacmid was a gift from Dr. Thomas C. Mettenleiter.
Antibody against UL37 was a gift from Dr. Weiming Yuan. Antibodies against GST (Z-5), IRF3 (FL-425), TRAF6 (D10) and RIG-I (H-300) were purchased from Santa Cruz Biotechnology. Antibodies against FLAG (M2, Sigma), V5 (A190-220A, Bethyl Group), RIG-I (SS1A, Enzo Life Sciences), STING (ab92605, Abcam), dsRNA-J2 (SCICONS), Sendai Virus (PD029, MBL), P-S172 TBK-1 (D52C2, Cell Signaling) and β-actin (Ab8226, Abcam) were purchased from the indicated suppliers. The glutamine analog 6-Diazo-5-oxo-L-norleucine (DON) was purchased from Sigma. Low molecular weight Poly [I:C] (31852-29-6), ppp-dsRNA (tlrl-3prna) and control-dsRNA (tlrl-3prnac) were purchased from InvivoGen Lipofectamine 2000 was purchased from Life Technologies.
For plasmid transfection in HEK293T cells, calcium phosphate transfection method was applied. 293T cells were plated at around 50%-60% confluence. For dsRNA and Poly [I:C] transfection in 293T cells and plasmid transfection in HeLa cells, Lipofectamine 2000 transfection reagent was used according to the manufacturer's instructions. Both cells were prepared at around 80%-90% confluence prior to transfection.
Lentiviruses were produced as previously described (Dong and Feng, 2011; Feng et al., 2008). Briefly, HEK293T cells were transfected with the packaging plasmids VSV-G and DR8.9 and the pCDH lentiviral expression vector or lentiviral shRNA plasmids. At 48 h post transfection, supernatant was harvested and filtered (and concentrated by centrifugation if necessary). HEK293T cells, MEFs, HeLa, HCT116 or HFF cells were infected with the supernatant in the presence of polybrene (8 μg/ml) with centrifugation at 1800 rpm for 45 minutes. Cells were selected at 48 h post infection and maintained in 10% FBS DMEM supplemented with puromycin (1˜2 μg/ml).
HEK293T cells, seeded in 24-well plates (˜50% cell density), were transfected with IFN-β, PRDIII (ISRE) or NF-κB reporter plasmid cocktail (50 ng of luciferase reporter plasmid and 5 ng of pRL Renilla luciferase control vector) and expression plasmid (empty plasmid, one or multiple plasmids depending on the experiment) by calcium phosphate precipitation. Cells were infected with SeV (100 HA/ml), HSV-1 for 16 h, transfected with Poly [I:C] for 16 h or directly harvested 30-36 h post transfection. Whole cell lysates were used to determine the activity of firefly luciferase and renilla luciferase by a microplate reader (FLUOstar Omega).
HSV-1 and VSV titer were determined by plaque assay on Vero monolayer essentially as previously described (Lieber and Bailer, 2013). Briefly, 10-fold serially-diluted virus-containing supernatant was added onto Vero cells and incubated for 2 h at 37° C. Then, DMEM containing 2% FBS and 1% methylcellulose (Sigma) was added after removing the supernatant. Plaques were counted at day 3 post-infection.
HFF cells were infected with HSV-1 for 8 h (MOI=50). HeLa cells were transfected with expression plasmid containing UL37 and subsequently infected with Sendai Virus for 6 h (200 HA/ml). Cells were fixed, permeabilized, stained with indicated primary antibody (1:100 dilution) and Alexa Fluor 488/594-congugated goat secondary antibody (1:200 dilution), and analyzed with confocal microscope (Leica). Representative images were shown for all analyses.
HEK293T cells were transfected with expression vector containing Flag-tagged gene of interest. Cells were harvested and lysed with Triton X-100 buffer (20 mM Tris, pH 7.5, 150 mM NaCl, 1.5 mM MgCl2, 20 mM β-glycerophosphate, 1 mM sodium orthovanadate, 10% glycerol, 0.5 mM EGTA, 0.5% Triton X-100) supplemented with a protease inhibitor cocktail (Roche). Whole cell lysates were sonicated and centrifuged at 12,000 rpm for 15 min. Supernatant was harvested, filtered, pre-cleared with protein A/G agarose beads at 4° C. for 1 h and then incubated with anti-Flag agarose beads at 4° C. for 4 h. The agarose beads were washed extensively and eluted with 0.2 mg/ml 3× Flag peptide. The eluted proteins were analyzed by SDS gel electrophoresis and silver staining.
For recombinant protein expression and purification, E. coli B121(DE3) was transformed with pGEX-4T-1 or pET28 plasmid containing UL37. Recombinant GST-UL37 expression was induced by 0.1 mM IPTG at 20° C. Bacteria were harvested, lysed and incubated with glutathione sepharose 4B (GE) for 4 h at 4° C. Sepharose beads were washed extensively and GST-UL37 was eluted with 10 mM reduced glutathione. UL37 was then cleaved and purified from the fusion protein by TEV protease treatment at 4° C. overnight.
For Co-IP using exogenous protein, HEK293T cells were transfected with indicated expression plasmids for 48 h. For Co-IP using endogenous proteins, cells were directly harvested. Whole cell lysates were prepared with NP40 buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% NP-40, 5 mM EDTA) supplemented with 20 mM β-glycerophosphate and 1 mM sodium orthovanadate. Whole cell lysates were sonicated, centrifuged and pre-cleared with protein A/G agarose for 1 h. Pre-cleared samples were then incubated with indicated antibodies overnight and protein A/G agarose for 1 h at 4° C., or with antibody/glutathione-conjugated agarose for 4 h at 4° C. The agarose beads were washed extensively and samples were eluted by boiling at 95° C. for 10 min. Precipitated proteins were analyzed by SDS gel electrophoresis and immunoblotting.
All immunoblottings were performed using the indicated primary antibodies (1:1000 dilution) and IRDye800-conjugated secondary antibodies (1:10,000 dilution, Licor). Proteins were visualized by Odyssey infrared imaging system (Licor).
Virus-infected HEK293T/Flag-RIG-I or HeLa/Flag-RIG-I stable cells were harvested and lysed in cold Triton X-100 buffer (20 mM Tris, pH 7.5, 150 mM NaCl, 1.5 mM MgCl2, 20 mM β-glycerophosphate, 1 mM sodium orthovanadate, 10% glycerol, 0.5 mM EGTA, 0.5% Triton X-100, 1 mM PMSF and 10 μg/ml leupeptin). Centrifuged supernatant was filtered and subjected to incubation with anti-Flag-conjugated agarose beads for 2 h at 4° C. Beads were then extensively washed and proteins were eluted with 3× Flag peptide at 0.2 mg/ml.
Gel filtration with superose 6 was performed as described previously. Briefly, eluted proteins (200-300 μl) were loaded to superose 6 column and subjected to gel filtration analysis with Buffer B (20 mM Tris-HCl, pH 7.6, 150 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 0.5% Triton X-100, 20 mM NaF, 20 mM β-glycerophosphate, 1 mM Na3VO4, 2.5 mM metabisulphite [sodium salt], 5 mM benzamidine). Elution was collected in 0.5 ml fractions and aliquots of fractions were analyzed by immunoblotting.
Mock- or HSV-1-infected cells (2×107) were harvested and lysed in 300 μl cold Triton X-100 buffer. Samples were sonicated briefly and centrifuged. Supernatant was filtered and loaded to superose 6 column and subjected to gel filtration analysis with Buffer B. Elution was collected in 0.5 ml fractions and aliquots of fractions were analyzed by immunoblotting.
Quantitative Real-Time PCR (qRT-PCR)
Quantitative Real-time PCR was performed as previously described. Cells were infected or treated with viruses or agents for indicated time period. Total RNA was extracted using TRIzol reagent (Invitrogen). Complementary cDNA was synthesized from DNase I-treated total RNA using reverse transcriptase (Invitrogen). cDNA was diluted and qRT-PCR was performed using SYBR Green Master Mix (Applied Biosystems) by real-time PCR instrument (Applied Biosystems). Relative mRNA expression for each target gene was calculated by the 2−ΔΔCt method using β-Actin as an internal control. The sequences of qRT-PCR primers are as follows:
Commercial cytokine ELISA kits used in this study include: human IFN-β (PBL Assay Science) and human RANTES (R&D Systems). Cytokine levels in the supernatant from cultured cells were assessed according to manufacturer's instruction. Absorbance was determined with FLUOstar Omega (BMG Labtech.).
Purified RIG-I or RIG-I mutants were incubated with 5′-ppp-dsRNA (Invivogen) at 37° C. for 20 min in ATPase reaction buffer (50 mM Tris-HCl, pH 7.5, 2.5 mM MgCl2, and ATP. Released phosphates were measured using a PiColorLock™ phosphate detection reagent (Innova Biosciences). For reactions with varying concentrations of ATP, the concentrations of RIG-I proteins and RNA were 20 nM and 80 nM, respectively. For reactions with varying concentrations of the RNA, the concentrations of RIG-I proteins and ATP were 20 nM and 500 μM, respectively.
For identification of deamidation sites, HEK293T/Flag-RIG-I stable cell line was transfected with an expression plasmid containing UL37 or infected by HSV-1 for 10 h (MOI=10). Flag-RIG-I was purified by anti-Flag-conjugated agarose beads for 4 h at 4° C. Beads were then extensively washed and RIG-I was eluted with 3× Flag peptide at 0.2 mg/ml. Purified RIG-I was subjected to SDS page electrophoresis and gel slices were prepared for in-gel digestion and Mass Spectrometry analysis (Harvard Taplin Mass Spectrometry Facility).
For Cysteine labeling experiment, bacterial purified UL37 (571-1123) was treated with N-methylacetamide (Alfa Aesar) (1 μM) at room temperature for 45 min. Samples were then blocked with Iodoacetamide (Sigma) (50 mM) at room temperature for 1 h and subjected to Mass Spectrometry analysis (Poochon Scientific).
Statistical analysis was performed by unpaired two-tailed Student's t-test. *, p<0.05; **, p<0.01; ***, p<0.001. A p-value less than 0.05 is considered statistically significant.
Applicant previously reported that the vGAT proteins of human KSHV and murine γHV68 recruit PFAS to deamidate RIG-I. In addition to these gamma herpesviruses, HSV-1 infection also increased negative charge of RIG-I as analyzed by two-dimensional gel electrophoresis (2-DGE), indicative of deamidation (
To probe the roles of RIG-I in host defense against HSV-1, Applicant depleted RIG-I expression and assessed the IFN-β mRNA in primary human foreskin fibroblasts (HFF). Applicant found that knockdown of RIG-I reduced IFN-β mRNA at 24 h induced by HSV-1 infection (
To determine whether HSV-1 infection inhibits RIG-I activation, Applicant sequentially infected 293T cells with HSV-1 and SeV, and determined IFN-β and ISG56 expression. Applicant found that HSV-1 infection significantly reduced IFN-β and ISG56 mRNA induced by SeV (
HSV-1 UL37 Interacts with RIG-I
To delineate the mechanism by which HSV-1 abrogates RIG-I activation, Applicant screened for RIG-I-binding proteins by co-immunoprecipitation (Co-IP) using a HSV-1 expression library, with a particular focus on gene products that operate in the early phase of infection. Co-IP assays identified open reading frames UL21 and UL37 as RIG-I-interacting proteins (
Applicant established 293T cells stably expressing UL37 (
To probe the effect of UL37 on signaling events downstream of RIG-I, Applicant analyzed the phosphorylation of TBK-1 (Ser172) and IRF3 (Ser396), markers of activated TBK-1 and IRF3, respectively. As shown in
HSV-1 infection reduced the charge of RIG-I, suggesting that HSV-1 induces RIG-I deamidation. Applicant found that UL37 expression was sufficient to reduce the charge of RIG-I, but not that of R-actin (
To probe the mechanism of UL37-induced deamidation, Applicant first determined whether a specific inhibitor of glutamine amidotransferase, 6-diazo-5-oxo-L-norleucine (DON), can block UL37-induced RIG-I deamidation. Indeed, DON inhibited RIG-I deamidation in cells expressing UL37 (
Applicant previously showed that m-vGAT induced deamidation and concomitant activation of RIG-I. However, the RIG-I-DD mutant failed to activate NF-κB and IFN-β reporters (
To assess the functional consequence of RIG-I deamidation, Applicant examined RIG-I activation by gel filtration. SeV infection induced oligomerization of RIG-I-WT as evidenced by fractions corresponding to protein complexes of ˜440-670 kDa sizes, while RIG-I-WT in mock-infected cells eluted in fractions corresponding to ˜130-230 kDa (
Applicant's mutational analysis indicates that N549 is critical for the RNA-binding and ATPase activities of RIG-I. Previously solved crystal structure of RIG-I showed that the amide group of N549 (within α24) forms two hydrogen bonds with the backbone of threonine 504 of the RNA-binding α-helix (α23) (
Applicant reasoned that only the deamidation-resistant RIG-I-QQ mutant will confer gain-of-function in RIG-I-mediated innate immune response, thus Applicant used wild-type HEK293 to establish stable cell lines expressing RIG-I wild-type and mutants. In resting cells, the level of phosphorylated TBK-1 (Ser172) was below detection in all four cell lines. HSV-1 infection increased the phosphorylation of TBK-1 to similar levels in control cells and cells expressing RIG-I-WT or RIG-I-DD (
UL37 purified from E. coli is sufficient to deamidate RIG-I, implying that UL37 is a bonafide protein deamidase. Because all known protein deamidases (e.g., PFAS) are cysteine hydrolases (Zhao et al., 2016), Applicant suspect that UL37 also contains a catalytic cysteine residue. Thus, Applicant mutated all 14 cysteines of UL37 individually to serines and screened for the loss of inhibition of RIG-I-mediated activation of the PRDIII promoter upon SeV infection. The C819S and C850S mutants were identified to have greatly impaired blockade of PRDIII induction by SeV (
To pinpoint the cysteine residue of the active site. Applicant employed a small molecule electrophile for mass spectrometry analysis, an approach that was successfully used to quantitatively profile functional cysteines in proteomes (Weerapana et al., 2010). The rationale is that functional cysteines, such as those in enzymatic active sites, are hyper-reactive and react with small molecule electrophiles independent of concentration. As such, a ratio of the percentage of labeled peptides at high concentration to that at low concentration near 1 predicts functional cysteines. After reacting with 2-Chloro-N-(hydroxymethyl) acetamide (CNM), mass spectrometry analysis identified that C819 was primarily labeled by CNM within UL37C. Specifically, 38.3° % and 42.5% of C819 were labeled by CNM at 1 and 10 μM, respectively (
To probe the roles of UL37-mediated deamidation in viral infection, Applicant introduced UL37 wild-type (UL37-WT) and UL37-C819S into the HSV-1 genome (designated HSV-1 UL37-WT and HSV-1 UL37-C819S) and examined RIG-I-mediated innate immune signaling. Gel electrophoresis of viral genomic DNA after BamHI digestion revealed identical pattern of migration, indicative of lack of large chromosome rearrangement (
Applicant then analyzed HSV-1 lytic replication and found that HSV-1 UL37-C819S produced ˜10% of virion progeny of HSV-1 UL37-WT in HFF (
Applicant previously reported that vGAT pseudo-enzymes of human KSHV and murine γHV68 recruited cellular PFAS to deamidate RIG-I and evade antiviral cytokine production (He et al., 2015). Interestingly, HSV-1 infection also induced RIG-I deamidation, despite the fact that genomes of alpha herpesviruses do not contain sequence homologues of vGAT proteins. Herein, Applicant identified UL37 as a viral deamidase that targets RIG-I for deamidation and inactivation, thereby preventing RIG-I from sensing viral dsRNA. To Applicant's knowledge, this is the first viral protein deamidase identified thus far. Previously reported protein deamidases contain either a cysteine-protease fold or a GAT domain (Cui et al., 2010; He et al., 2015; Sanada et al., 2012; Wang et al., 2009). UL37-mediated deamidation of RIG-I disarms downstream innate immune signaling, suggesting the critical, and likely more ubiquitous, roles of protein deamidation in signal transduction. UL37 is a large tegument protein that is implicated in viral trafficking, egress and innate immune regulation (Desai et al., 2001; Liu et al., 2008; Pitts et al., 2014). Taken together, UL37 inhibits the IRF-IFN branch of innate immune signaling through deamidation of RIG-I, while activating the NF-κB cascade, sharing functions similar to the gamma herpesvirus vGAT proteins.
Applicant's biochemical analyses show that UL37 is intrinsically a protein deamidase. UL37 and its carboxyl terminal fragment (571-1123) purified from E. coli were sufficient to deamidate RIG-I in vitro. Mutational analysis and electrophile reaction profiling of hyper-reactive cysteines identified C819 as the single residue critical for the deamidase activity, implying that C819 is the active cysteine of the catalytic triad of UL37. Interestingly, C850 is more conserved in alpha herpesviruses than C819 (data not shown). The fact that C850 is largely inaccessible suggests that it may be required for the structural integrity of the deamidase domain. It is unclear whether other UL37 homologs are deamidases. Future structural studies of the UL37 deamidase domain may define a new fold catalyzing protein deamidation and “visualize” the catalytic cysteine.
Although previous studies implicated RIG-I in sensing dsRNA produced by herpesviruses (da Silva and Jones, 2013; Jacquemont and Roizman, 1975; Rasmussen et al., 2009; Weber et al., 2006), Applicant's work provides further credence concerning the RIG-I-mediated immune defense against a model DNA virus and viral immune evasion thereof. HSV-1 infection prevents RIG-I activation and innate immune responses triggered by subsequent SeV infection. These phenotypes were recapitulated by UL37 expression, pointing to the key roles of UL37 in evading RIG-I activation by viral dsRNA. The deamidated RIG-I-DD (D495 and D549) mutant, failed to sense 5′ppp-RNA and SeV, which correlated with its inability to initiate host immune signaling and control VSV replication. Comparing HSV-1 replication kinetics in IFN-competent 293T and HeLa cells to that in IFN-deficient Vero cells, Applicant found that the deamidase activity of UL37 is critical in negating RIG-I-mediated inhibition of the early steps of HSV-1 lytic replication. The mutation abolishing UL37 deamidase activity, notably, also impaired HSV-1 replication during late stages of replication in an RIG-I-independent manner, implying the existence of other viral and cellular targets in addition to RIG-I. Nevertheless, uncoupling RIG-I deamidation from UL37, via either introducing the deamidation-resistant RIG-I-QQ into cells or engineering the C819S mutation of UL37 into the HSV-1 genome, restored RIG-I activation and downstream innate immune signaling, thereby reducing HSV-1 productive infection. These results unambiguously demonstrate the antiviral activity of RIG-I against a DNA herpesvirus and elucidate a new mechanism of viral immune evasion.
N495 and N549 reside in two α-helices that constitute the RNA-binding interface of the Hel2i domain. Interestingly, N549 forms hydrogen bonds with the backbone of T504 that ends the N495-containing α23 helix, providing a physical link between these two neighboring helices that are located immediately proximal to the RIG-I-bound dsRNA. These observations suggest that the two α-helices constitute a region responsible for regulating RNA-binding/sensing by RIG-I. The susceptibility of the hydrogen bonds between N549 and T504 to the deamidase activity of UL37 underpins the inactivation of RIG-I by HSV-1 infection. Remarkably, the N549Q mutation appears to conserve hydrogen bonds, and confers resistance to UL37-mediated deamidation, demonstrating the exquisite specificity of UL37-mediated deamidation. Deamidation of N495 and N549 within the Hel2i domain, unexpectedly, abolishes 5′ppp-RNA-binding and ATP hydrolysis of RIG-I, uncovering a simple but powerful mechanism to switch off RIG-I. Although the CTD of RIG-I is responsible for sensing viral dsRNA, emerging studies support the regulatory role of helicase domains in RNA-sensing by RIG-I. It was previously reported that Hel2i “measures” the length of dsRNA stem during RNA-binding by RIG-I (Kohlway et al., 2013). Structural analysis also highlighted the direct contact between Hel2i and dsRNA (Kowalinski et al., 2011; Luo et al., 2011). Moreover, mutations within a helicase domain reduced the ATPase activity of RIG-I, increased its association with cellular dsRNA and activated downstream signaling (Lassig et al., 2015). Together with these observations. Applicant's work further lends credence to the pivotal roles of Hel2i of RIG-I and site-specific deamidation thereof in interacting with and sensing viral dsRNA, suggesting more ubiquitous roles of protein deamidation in fundamental biological processes.
Cells (1×106) were lysed in 150 μl rehydration buffer (8 M Urea, 2% CHAPS, 0.5% IPG Buffer, 0.002% bromophenol blue) by three pulses of sonication and whole cell lysates were centrifuged at 20,000 g for 15 min. Supernatants were loaded to IEF strips for focusing with a program comprising: 20 V, 10 h (rehydration); 100 V, 1 h; 500 V, 1 h; 1000 V, 1 h; 2000 V, 1 h; 4000 V, 1 h; 8000 V, 4 h. After IEF, strips were incubated with SDS equilibration buffer (50 mM Tris-HCl [pH8.8], 6 M urea, 30% glycerol, 2% SDS, 0.001% Bromophenol Blue) containing 10 mg/ml DTT for 15 min and then SDS equilibration buffer containing 2-iodoacetamide for 15 min. Strips were washed with SDS-PAGE buffer, resolved by SDS-PAGE, and analyzed by immunoblotting.
GST-RIG-I was purified from transfected 293T cells to homogeneity as determined by silver staining. In vitro on-column deamidation of RIG-I was performed as previously reported (He et al., 2015). Briefly, ˜0.2 μg of His-tagged UL37/UL37 (571-1123) expressed and purified from E. coli, and 0.6 μg of GST-RIG-I (bound to glutathione-conjugated agarose) were added to a total volume of 30 μl. The reaction was carried out at 30° C. for 45 min in deamidation buffer (50 mM Tris-HCl, pH 7.5, 100 mM NaCl, 5 mM MgCl2). Protein-bound GST beads were washed with deamidation buffer and GST-RIG-I was eluted with rehydration buffer (6 M Urea, 2 M Thio-urea, 2% CHAPS, 0.5% IPG Buffer, 0.002% bromophenol blue) at room temperature. Samples were then analyzed by two-dimensional gel electrophoresis and immunoblotting.
Recombinant HSV-1 was engineered as previously described (Dong et al., 2010). Briefly, DNA fragments containing UL37 WT and C819S were amplified using overlapping primers. First round PCR products of ˜500 bp fragment upstream of UL37, UL37 open reading frames (WT and C819S) and ˜500 bp fragments downstream of UL37 were used as the template for second round PCR amplification. Purified PCR products of the second round, along with HSV-1 ΔUL37 (KOS) Bacmid, were transfected into 293T cells to generate recombinant HSV-1. The revertant (containing wild-type UL37, designated wild-type) and UL37-C819S mutant were plaque purified and validated by restriction digestion of viral genomic DNA and sequencing of the UL37 open reading frame.
RNA EMSA was performed as previously described (Takahasi et al., 2008). 5′-ppp-dsRNA and control dsRNA were purchased from Invivogen and bottom strands were labeled with γ-[P32]ATP by T4 polynucleotide kinase (NEB). Purified RIG-I and RIG-I mutants were incubated with dsRNA at room temperature for 15 min. Binding buffer contains 20 mM Tris-HCl (pH=8.0), 1.5 mM MgCl2 and 1.5 mM DTT. Unlabeled ppp-dsRNA was used as competitor at 500-fold in excess. The reaction mixtures were run on 5% native polyacrylamide gels at a constant voltage of 200 V. Gels were dried and subjected to phosphorimaging.
Purified RIG-I or RIG-I mutants were incubated with 5′-ppp-dsRNA (Invivogen) at 37° C. for 20 min in ATPase reaction buffer (50 mM Tris-HCl, pH 7.5, 2.5 mM MgCl2, and ATP). Released phosphates were measured using a PiColorLock™ phosphate detection reagent (Innova Biosciences). For reactions with varying concentrations of ATP, the concentrations of RIG-I proteins and RNA were 20 nM and 80 nM, respectively. For reactions with varying concentrations of the RNA, the concentrations of RIG-I proteins and ATP were 20 nM and 500 μM, respectively.
Upon infection, eukaryotic cells immediately respond with innate immune activation to defeat the invading pathogens. Cyclic GMP-AMP (cGAMP) synthase (cGAS) is an essential cytosolic sensor that detects double-stranded (ds) DNA of microbial origin or aberrantly localized cellular DNA. Other DNA sensors, including AIM2, DAI, DDX41, RNA polymerase III, DNA-PK and IFI16, may play redundant roles in a tissue- or ligand-specific manner in detecting cytosolic dsDNA. Upon binding dsDNA, cGAS catalyzes the synthesis of cGAMP, which induces the dimerization and activation of the ER-anchored STING (also known as MITA). Within close proximity to the ER membrane, STING recruits TBK-1 and interferon regulatory factor 3 (IRF3) to assemble into a signaling complex that phosphorylates and activates IRF3. Along with NF-κB and AP-1, nuclear IRF3 potently up-regulates the gene expression of interferons (IFNs). IFNs, via autocrine and paracrine mechanisms, stimulate the expression of hundreds of genes, known as ISGs, which establish an immune defensive state of the cell. Parallel to the TBK-1-IRF3-IFN pathway, IKK kinase, consisting of IKKα, IKKβ and IKKγ (also known as NEMO), phosphorylates and induces the degradation of inhibitor of NF-κB (IκB). This enables NF-κB activation that induces the expression of inflammatory cytokines, such as interleukins and chemokines. The primary role of inflammatory cytokines is to attract professional immune cells to the site of infection. Thus, the innate immune system defends the host from infection via direct anti-microbial activities and enables the establishment of adaptive immunity in tissue local to the infection.
Though key steps of the cGAS-STING pathway are well established, the regulatory mechanisms governing cGAMP synthesis of cGAS to induce STING-dependent innate immune activation is not well understood. Studying viral immune evasion allows us to interrogate mechanisms regulating host immune responses. As one of the most successful pathogens, herpesviruses have evolved numerous intricate strategies to manipulate, evade and exploit host immune response to benefit their infection. The most common viral mechanism is to encode proteins that physically interact with central cellular signaling nodes of immune defense to derail host immune response. Viral proteins efficiently regulate cellular immune signal transduction by microbial enzyme-mediated reactions, such as proteolytic cleavage or post-translational modifications (PTMs). Virally encoded proteases cleave various adaptor molecules and effectively dampen innate immune signaling, while host cells often deploy reversible PTMs (phosphorylation, ubiquitination and sumoylation) to regulate immune response.
Protein deamidation is emerging as a key PTM that regulates immune responses against infecting microbes. First reported more than half a century ago, protein deamidation was regarded as a marker associated with protein “aging” or functional decay. Though initial studies focused on non-enzymatic protein deamidation, recent findings from bacterial effectors and mammalian cells imply that protein deamidation can be enzyme-catalyzed and thus highly regulated. Applicant has identified viral pseudo-enzymes and bona fide deamidases that target cellular innate immune RIG-I sensor to evade antiviral cytokine roduction. While gamma herpesvirus vGAT pseudoenzymes recruit cellular PFAS to deamidate RIG-I, the UL37 tegument protein of herpes simplex virus 1 (HSV-1) is a bona fide protein deamidase that deamidates RIG-I in vitro and in cells. While further characterizing the in vivo roles of UL37 deamidase in HSV-1 infection, Applicant discovered that UL37 antagonizes cGAS-mediated innate immune activation via deamidating cGAS. Moreover, HSV-1 carrying deamidase-deficient UL37 was highly attenuated, and more robustly induced innate and adaptive immune responses in mice than wild-type HSV-1. Vaccination with HSV-1 carrying deamidase-deficient UL37 protected mice from lethal dose challenge with wild-type HSV-1. These results imply that interfering with protein deamidation can boost antiviral immune responses and thwart viral infection.
Applicant reports herein that the UL37 tegument protein of HSV-1 deamidates RIG-I to avoid dsRNA-induced innate immune activation. Recombinant HSV-1 carrying deamidase-deficient UL37C819S mutant (HSV-1 UL37C819S) more robustly induced antiviral cytokines than HSV-1 containing wild-type UL37 (HSV-1 UL37WT). To further characterize this recombinant HSV-1, Applicant examined antiviral immune responses in human THP-1 monocytes upon HSV-1 infection. Real-time PCR analysis of representative antiviral cytokines (IFNB1, ISG56, CXCL10, MX1, IFIT3 and IL6) indicated that HSV-1 UL37C819S virus induced ˜5-10-fold higher expression of cytokine genes than HSV-1 UL37 wild-type (WT) in THP-1 cells during early viral infection (
cGAS is a crucial DNA sensor that detects cytosolic DNA of diverse human pathogens, including herpesviruses. Thus, Applicant assessed whether cGAS is required for effective antiviral immune responses against HSV-1 UL37C819S virus. Applicant infected wild-type and cGAS-deficient L929 fibroblasts with HSV-1 UL37WT and HSV-1 UL37C819S, and determined antiviral gene expression. In wild-type L929 fibroblasts, HSV-1 UL37C819S virus more robustly induced Isg56 and Cxcl10 expression than HSV-1 UL37WT virus, recapitulating the phenotype that was observed in human THP-1 monocytes. Remarkably, loss of cGAS abolished Isg56 and Cxcl10 expression in response to HSV-1 UL37WT and HSV-1 UL37C819S (
To probe the effect of HSV-1 infection on the DNA-cGAS pathway, Applicant determined intracellular cGAMP concentrations using the THP-1/Lucia reporter cell line. Applicant applied known concentrations of cGAMP to establish a standard that demonstrated a high correlation between cGAMP concentration and luciferase activity with 0-30 ng/ml of cGAMP. Applicant determined that HSV-1 UL37WT induced approximately 3.5 ng of cGAMP per one million of THP-1 cells, while HSV-1 UL37C819S infection increased cGAMP production to ˜10.5 ng per one million of THP-1 cells (
UL37 Targets cGAS to Dampen Antiviral Cytokine Production
To determine whether UL37 is sufficient to inhibit cGAS-mediated innate immune responses, Applicant established a THP-1 cell line stably expressing UL37 by lentiviral transduction (
Given that recombinant HSV-1 UL37C819S virus more robustly induced antiviral cytokine production in THP-1 cells (
UL37 Deamidates cGAS In Vitro and in Cells
UL37WT, but not the deamidase-deficient UL37C819S mutant, reduced cGAS-mediated cGAMP synthesis. Moreover, HSV-1 UL37C819S virus more robustly induced antiviral cytokines in THP-1 monocytes than HSV-1 UL37WT. These results imply that UL37 targets cGAS for deamidation. To test this hypothesis, Applicant first determined whether UL37 interacts with cGAS in HSV-1-infected cells. Using recombinant HSV-1 carrying FLAG-tagged UL37, Applicant demonstrated that cGAS precipitated with UL37 in HSV-1-infected THP-1 cells (
To assess whether UL37 induces cGAS deamidation, Applicant analyzed the charge status of cGAS without or with UL37 expression by two-dimensional gel electrophoresis (2DGE). As shown in
Applicant has previously shown herein that UL37 is a bona fide protein deamidase of RIG-I. Thus, Applicant sought to determine whether UL37 is sufficient to deamidate cGAS in vitro. Applicant purified cGAS, UL37WT and UL37C819S mutant from bacteria to high homogeneity (
Deamidation Impairs the cGAMP Synthase Activity of cGAS
To probe the role of protein deamidation in cGAS-mediated antiviral immune response, Applicant first performed reporter assays to analyze the ability of various deamidated cGAS mutants in activating the IFN-β and NF-κB promoters. Applicant also has also generated mutations of all N and Q residues that are conserved within the Mab21 enzyme domain of human and mouse cGAS for these reporter assays. These reporter assays showed that N210D reduced cGAS-mediated gene expression by 50%, while the other three deamidations had marginal effects. The other deamidated residues did not significantly impair cGAS to activate the IFN-β promoter. However, combining the three mutations in NQQ389,451,454DEE modestly reduced cGAS-induced gene expression. When all four deamidated residues were introduced into cGAS, Applicant found that the cGAS-DDEE mutant failed to activate the IFN-β and NF-κB promoters by reporter assay (
All four deamidation sites, N210, N389, Q451 and Q454, are conserved between mouse and human cGAS. These four sites correspond to N196, N377, Q436 and Q439 of mouse cGAS (mcGAS). Previous structural studies revealed an active site of mcGAS that catalyzes the synthesis of cGAMP, consisting of two parallel β-sheets (32 and 37, PDB: 4K9B) (
Interestingly, this hydrophobic pocket is formed by residues from core β-sheets (F212, V214 and F216 of β2, V306 and 1308 of 07) and a neighboring α-helix (V171, L175 and L179 of α2). In the cGAS structure bound to dsDNA, N196 lies between the hydrophobic cluster and the backbone of the dsDNA†(PDB:4K9B). Moreover, structural analysis by others show that, similar to other nonpolar residues with small side chain, N196 (or N210 of hcGAS) confers flexibility to the activation loop of cGAS. Thus, deamidation of N196 of mcGAS is expected to impinge on the nearby hydrophobic cluster and the flexibility of the activation loop that collectively enable the proper coordination of the catalytic triad.
This is supported by the structure wherein N196 is close to the catalytic residue D213. In fact, the three catalytically residues E211, D213 and D307 form a highly negatively charged spot on protein, whose structure and physical chemical properties are likely very sensitive to alternation of nearby electrostatic potential induced by the damindation of N196. Applicant therefore assessed the effect of N196 deamidation on the enzyme activity of cGAS. As shown in
Structural analyses also indicate that the side chain of N376 and N377 of mcGAS (corresponding to N388 and N389 of hcGAS) project toward the minor groove of the dsDNA helix, suggesting that deamidation of these residues potentially interferes cGAS ability to sense dsDNA. However, precipitation of biotinylated interferon-stimulating DNA (ISD) demonstrated that neither UL37-WT, nor UL37C819S diminished cGAS co-precipitated with ISD. In fact, UL37WT, but not UL37C819S, increased the interaction between cGAS and ISD by ˜50%. Similar results were recapitulated with the deamidated cGAS-DDEE mutant, which demonstrated slightly enhanced interaction with ISD. Given that all four deamidation sites reside in regions proximal to the dimerization interface of cGAS, Applicant sought to determine whether UL37 influences cGAS self-dimerization. Co-IP assay showed that expression of UL37WT or UL37C819S did not alter cGAS dimerization. Taken together, these results suggest that UL37-mediated deamidation does not impair either the dsDNA-binding or dimerization of cGAS.
Deamidated cGAS Fails to Activate Innate Immune Signaling and Restrict DNA Virus Replication
To probe the role of deamidation in regulating cGAS-mediated immune signaling and restricting viral replication, Applicant “reconstituted” cGAS-deficient L929 cells with cGAS wild-type and the deamidated cGAS-DDEE mutant (
When infected with HSV-1 UL37WT or HSV-1 UL37C819S virus, L929 cells “reconstituted” with cGAS wild-type up-regulated the expression of inflammatory genes as potent as wild-type L929 cells. L929 cells “reconstituted” with cGAS-DDEE essentially behaved like cGAS-deficient L929 cells, demonstrating no induction of immune gene expression in response to HSV-1 infection (
To characterize the in vivo function of the deamidase activity of UL37, Applicant infected mice with HSV-1 UL37WT and HSV-1 UL37C819S virus. At 8 hours post-infection, HSV-1 UL37C819S virus induced ˜2-5-fold more cytokines in the sera of infected mice than HSV-1 UL37WT (
Previous studies have implicated the cGAS-STING pathway in promoting adaptive immune responses. Thus, Applicant tested whether the increased innate immune activation by HSV-1 UL37C819S virus translated into enhanced adaptive immunity. To quantify T cell immunity, Applicant analyzed virus-specific CD8+ T cells by tetramer staining against the most abundant epitope of glycoprotein B (gB, 498-505, SSIEFARL). This analysis showed that both HSV-1 UL37WT and HSV-1 UL37C819S induced similar CD8+ T cell response kinetics, peaking at 6 days post-infection (dpi) (
To determine whether the elevated virulence of HSV-1 UL37WT is dependent on its ability to evade cGAS-mediated innate immune activation, Applicant analyzed the pathogenesis of HSV-1 UL37WT and HSV-1 UL37C819S in mice deficient in cGAS or STING. Mice deficient in cGAS or STING were highly susceptible to HSV-1 infection, demonstrating 100% lethality by 11 dpi. Importantly, cGAS-deficient mice infected with HSV-1 UL37C819S succumbed to death as rapidly as those infected with HSV-1 UL37WT (
Immunization with HSV-1 UL37C819S Protects Mice from HSV-1 Lethal Dose
Considering that HSV-1 UL37C819S more robustly induces immune responses and is highly attenuated in mice, Applicant explored the possibility that immunization with HSV-1 UL37C819S protects mice from pathogenesis induced by wild-type HSV-1 infection. For this experiment, Applicant used BALB/c mice, which are more susceptible to HSV-1 infection than BL/6 mice. After two rounds of HSV-1 UL37C819S infection at an interval of two weeks (
To further characterize the pathology of HSV-1 infection, Applicant analyzed the brain of mice infected with HSV-1. Haematoxylin & Eosin (H&E) staining showed a significant fraction of cells had apparent morphology changes only in mice immunized with PBS and challenged with wild-type HSV-1 (
As innate immunity is essential to defeating pathogen infection, pathogens have evolved diverse mechanisms to evade host defense, providing a physiological system to examine host immune regulation. Employing HSV-1 for monocyte and mouse infection, Applicant discovered that the UL37 tegument protein of HSV-1 deamidates cGAS to abrogate its cGAMP synthesis activity, without diminishing the DNA-binding or dimerization. Site-specific deamidation of all four amide-containing residues distributed throughout the relatively large enzyme domain reveals an exquisite specificity of deamidation on the enzyme activity of cGAS. The physiological role of the deamidase activity of UL37 in counteracting cGAS-mediated immune defense is substantiated by significantly elevated levels of inflammatory cytokines in THP-1 monocytes and mice infected with the deamidase-deficient HSV-1 UL37C819S than those infected with HSV-1 UL37WT. Applicant further showed that elevated antiviral cytokines translated into more robust adaptive immunity against HSV-1 in mice, including CD8+ cytotoxic T cell response and serum antibody. These findings agree with a previous report that cGAMP and activation of cGAS-mediated innate immune signaling play an adjuvant role in immunization. In support of this conclusion, immunization with the highly inflammatory deamidase-deficient HSV-1 UL37C819S that had attenuated replication in vivo protected mice from challenge with lethal dose of wild-type HSV-1, representing a new vaccine candidate.
Applicant has shown that UL37 deamidates RIG-I to prevent dsRNA-induced activation. This work identifies cGAS as an additional target of UL37 in HSV-1-infected cells. In cGAS- and STING-deficient mice infected with HSV-1, Applicant found that the deamidase-deficient HSV-1 UL37C819S virus was as pathogenic as wild-type HSV-1, as measured by survival rates of mice infected with HSV-1 UL37WT and HSV-1 UL37C819S. These results clearly support the crucial role of UL37 in antagonizing the cGAS-STING pathway, but do not address the role of UL37-mediated RIG-I deamidation, previously shown to diminish antiviral cytokine production, in host defense against HSV-1 infection in mice. The identical pathogenesis of HSV-1 UL37WT and HSV-1 UL37C819S virus in mice deficient in cGAS or STING suggests that UL37 fails to antagonize mouse RIG-I in vivo. N495 of human RIG-I is not conserved in mouse, so it is possible that mouse RIG-I is resistant to UL37-mediated deamidation and inhibition. Although the roles of RIG-I in HSV-1 infection in vivo remain undefined, RIG-I is possibly important for innate immune defense against HSV-1 in cell types with limited or minimal cGAS expression, such as epithelial cells and keratinocytes. Previous studies demonstrating the antiviral activities of RIG-I against various herpesviruses primarily used mouse fibroblasts or human cells deficient in RIG-I.
Remarkably, all four cGAS deamidation sites impinge on cGAMP synthesis activity despite being located within three structurally distinct surfaces of cGAS. Two structural studies highlighted the importance of the N210 of hcGAS (or N196 of mcGAS) in regulating cGAS enzymatic activity. Specifically, others showed that N210 is located within the first half of the so-called activation loop. The sequence of this short loop features residues that have small and non-charged side chains. Additional mutational and functional analysis of G211 and S212 of hcGAS in this structural study demonstrated that the flexibility of the activation loop underpins the conformational change and subsequent coordination of the catalytic triad of cGAS upon DNA-binding and dimer formation. Thus, deamidation of N210 of hcGAS is expected to compromise the free rotation of the activation loop and proper formation of the catalytic triad.
Surprisingly, collective deamidation of N389, Q451 and Q454 reduced cGAMP synthesis, but not DNA-binding and dimerization, of cGAS. N389 and N388 lie at the center of the dsDNA-binding surface of cGAS and directly point to the minor groove of dsDNA. Deamidation of N389, and more so that of N388, are expected to diminish the DNA-binding ability of cGAS. However, Applicant's reporter assay showed that N388D and N389D mutations had no detectable effect on the ability of cGAS to activate the IFN-β promoter. Moreover, UL37WT expression and the deamidated cGAS mutant (cGAS-DDEE) appeared to slightly increase the DNA-binding of cGAS. Q451 and Q454 reside in a short α-helical structure that forms the front edge of the butterfly-shaped cGAS dimer. The expression of UL37WT and UL37C819S mutant had no detectable effect on cGAS dimer formation upon HT-DNA transfection. These results indicate that deamidation of cGAS does not impair the DNA-binding and dimerization of cGAS upon sensing dsDNA. On the other hand, cGAS-deficient L929 cells “reconstituted” with cGAS mutants harboring single deamidated residues, demonstrated lower activity to induce Ifnb 1 expression in response to transfected HT-DNA, suggesting that these sites are important for cGAS signaling. Thus, Applicant's reported methods are perhaps not sufficiently sensitive to accurately quantify the dsDNA-cGAS interaction, especially given the observation that the DNA-binding of cGAS appears to be of low affinity. If indeed the deamidation of these Gin and Asn residues do not impair the dimerization and DNA-binding of cGAS, it is possible that the deamidated surfaces of monomeric or dimeric cGAS serve as binding sites for cellular factors that regulate cGAS enzymatic activity. For example, cGAS sensing of HIV DNA requires the PQBP1 cofactor for innate immune activation. Whether deamidation impacts cGAS interaction with its cofactors remains to be determined. Nevertheless, the conformational changes induced by deamidation of these residues likely impact the active site and reduce cGAMP synthesis by cGAS. The molecular details of how these deamidations affect cGAS enzyme activity calls for further investigation. It is clear that these deamidations dampen the cGAMP synthesis activity of cGAS with explicit specificity.
cGAS is a cytosolic DNA sensor crucial for innate immune defense and aberrant activation of cGAS can lead to autoimmune diseases. Thus, the cGAS activity is tightly regulated. PTMs, such as phosphorylation, glutamylation, sumoylation and ubiquitination, play important roles in regulating the activity of cGAS. Phosphorylation of hcGAS S305 by AKT potently inhibits the enzymatic activity of cGAS. Glutamylation of cGAS by the enzymes TTLL6 and TTLL4 dampens the DNA-binding and synthase activity of cGAS, while the removal of glutamylation by CCP5 and CCP6 enhances cGAS activity.
Similarly, TRIM38 targets cGAS for sumoylation to prevent its polyubiquitination and degradation that is facilitated by SENP2-mediated desumoylation. Interestingly, sumoylation of cGAS at different residues suppresses its DNA-binding, oligomerization and synthase activities, and desumoylation by SENP7 increases cGAS activity. Thus, the activity of cGAS is dynamically regulated by sumoylation and desumoylation during infection. In this study, Applicant provides evidence that the activity of cGAS can be modified through deamidation, adding another PTM to the dynamic and complex regulation of cGAS.
Due to its core function in innate immune response against microbes, cGAS is often targeted by diverse pathogens to prevent innate immune activation. Human kaposi's sarcoma-associated herpesvirus (KSHV) notably deploys three distinct molecules, ORF52, LANA (ORF73) and vIRF1 (K9), to disable cGAS and its downstream signaling. The E7 oncogene of human papillomavirus and E1A of adenovirus utilize a common L×C×E motif to antagonize the DNA-sensing of cGAS. Recently, the protease cofactor NS2B of Dengue virus was shown to promote the lysosomal degradation of cGAS, thereby suppressing the induction of type I interferon production in infected cells. Applicant found that HSV-1 UL37 tegument protein deamidates cGAS to block cGAMP synthesis, revealing an efficient means of antagonizing the cGAS-STING pathway. Applicant and others have reported that herpesviruses and bacteria deploy deamidation to modify key signaling molecules to manipulate host immune responses.
In conclusion, Applicant has identified multiple sites of deamidation within cGAS targeted by HSV-1 UL37 deamidase. Deamidation of cGAS specifically ablates the cGAMP synthesis activity of cGAS. HSV-1 containing the deamidase-deficient UL37C819S is highly attenuated in mice and more robustly induces antiviral cytokines. Immunization with the deamidase-deficient HSV-1 virus potently protects mice from lethal dose challenge of HSV-1 wild-type. Collectively, these studies provide evidence that deamidation modifies protein function.
The genetic data disclosed in this experiment shows that the cGAS and STING pathway is the primary target of UL37. Moreover, recombinant HSV-1 containing the deamidase-deficient UL37C819S is highly attenuated in mice, but induced much more robust antiviral immune response (innate arm and adaptive as well). Thus, these proteins are shown to be effective as a prophylactic vaccine. Indeed, mice vaccinated with the recombinant HSV-1 containing the deaminase-deficient UL37C819S potently protected mice from lethal dose infection of wild-type HSV-1.
Commercially available antibodies used for this study include mouse monoclonal FLAG M2 antibody (Sigma), mouse monoclonal V5 antibody and β-actin antibody (Abcam), Phospho-TBK1 (Ser172) antibody, Phospho-IRF3 (Ser396) antibody and cGAS (D1D3G) antibody (Cell signaling), TBK1 antibody (Bethyl), His-probe antibody (H-3) and IRF3 antibody (FL-425) (Santa cruz), APC rat anti-mouse CD8a and PE hamster anti-mouse CD3ε (BD Biosciences).
Major histocompatibility complex (MHC)/peptide tetramers for HSV-1 gB 498-505/Kb (SSIEFARL) conjugated to PE were obtained from the NIH Tetramer Core Facility (Emory University, Atlanta, Ga.).
HSV-1 UL37 and VHS polyclonal antibodies were generated by repeatedly immunizing rabbit with purified proteins.
HT-DNA and LPS (Sigma-Aldrich), 2′, 3′-cGAMP (InvivoGen), streptavidin agarose (Thermo Fisher), Amylose Resin (New England Biolabs), Ni-NTA His-Bind Resin (Novagen), TEV protease (Invitrogen).
Biotin labeled ISD-45 DNA was ordered from IDT. [α-P32]-ATP was ordered from Perkin Elmer. Lipofectamine 2000 was purchased from Invitrogen.
THP1-Lucia ISG reporter cells (InvivoGen) was kindly provided by Dr. Fanxiu Zhu (Florida State University). L929 and L929 cGAS knockout cells were provided by Dr. Fanxiu Zhu.
MHV68 virus was propagated in BHK21 cells as previously described. HSV-1 WT and UL37 C819S recombinant viruses were propagated using VERO cells. cGAS knockout mice and BALB/c mice were purchased from the Jackson laboratory. STING knockout mice were provided by Dr. Jae Jung (the University of California). Six to eight-week old, gender-matched mice were used for all experiments. All animal work was performed under strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of the University of Southern California.
For HSV-1 infection, BL/6, cGAS KO or STING KO mice were infected with 5×107 PFU of virus via intraperitoneal injection for survival curve analysis. For tetramer staining and antibody measurement, HSV-1 was reduced to 106 PFU per mouse.
For BALB/c mice, the mice were immunized with HSV-1 UL37 C819S virus (106) twice with an interval of two weeks and then challenged with HSV-1 (5×106) via intraperitoneal infection.
RNA Extraction and qRT-PCR
THP-1 or L929 cells were infected with HSV-1 (MOI=5) or stimulated with HT-DNA (2 μg/ml) or cGAMP (2 μg/ml) for 6 h unless specifically indicated otherwise. Cells were washed with cold PBS, and total RNA was extracted by using TRIzol Reagent (Invitrogen). RNA was digested with DNase I (New England Biolabs) to remove genomic DNA. One microgram of total RNA was used for reverse transcription with PrimeScript Reverse Transcriptase (Clontech) according to the manufacturer's instruction. Approximately 0.5% of the cDNA was used as template in each quantitative real-time PCR (qRT-PCR) reaction with SYBR master mix (Applied biosystems).
HEK293T cells in 24-well plates were transfected with a reporter plasmid mixture that contained 50 ng of the plasmid expressing IFN-β or NF-κB firefly luciferase reporter and 20 ng of the plasmid expressing TK-renilla luciferase reporter. At 30 h post-transfection, cells were harvested and cell lysates were prepared. Cell lysates were used for dual luciferase assay according to the manufacturer's instruction (Promega).
Immunoprecipitation was carried out as described previously. Briefly, THP-1 cells were infected with HSV-1 FLAG-UL37 recombinant virus (MOI=0.5) for 16 h. The cells were harvested and lysed with NP40 buffer (50 mM Tris-HCl [pH 7.4], 150 mM NaCl, 1% NP-40, 1 mM EDTA, 5% glycerol) supplemented with a protease inhibitor cocktail (Roche). Centrifuged cell lysates were pre-cleared with Sepharose 4B beads and incubated with FLAG-agarose at 4° C. for 4 h. The agarose beads were washed three times with lysis buffer and precipitated proteins were released by boiling with 1×SDS sample buffer at 95° C. for 5 min. The resolved samples were applied to immunoblot analysis.
Mouse MBP-cGAS fusion protein (151-522) was expressed in BL21 (DE3) and the bacteria were grown at 37° C. to an OD600 of 0.6. Then the cultures were cooled to 18° C. and protein expression was induced by adding 0.1 mM Isopropyl b-D-1-thiogalactopyranoside (IPTG) for 16 h. Cells were collected by centrifugation and lysed with lysis buffer (20 mM Tris-HCl [pH 7.4], 200 mM NaCl, 10% glycerol, 0.5% Triton X-100, 0.2 mg/ml lysozyme supplemented with protease inhibitor cocktail). Clarified lysates were mixed with amylose resin and incubated for 2 h at 4° C. The resin was washed extensively with lysis buffer and the recombinant proteins were eluted by 10 mM maltose.
For Mass spectrometry analysis, purified MBP-mcGAS was digested with TEV protease overnight at 4° C. and MBP proteins were depleted by incubation with Ni-NTA agarose.
UL37 or UL37C819S were purified as previously described. Briefly, HEK293T cells were transiently transfected with a plasmid containing UL37 or UL37C819S, harvested and lysed with lysis buffer (20 mM Tris (pH 7.4), 150 mM NaCl, 10% (vol/vol) glycerol, 0.5% Triton X-100, and 0.5 mM DTT) supplemented with a protease inhibitor cocktail (Roche), and lysates were precipitated with 20 μL of FLAG M2-conjugated agarose (Sigma). After extensive washing with lysis buffer, precipitated proteins was eluted with FLAG peptide (100 μg/ml) and used for in vitro deamidation assay.
Cells or in vitro deamidation samples were resolved in 150 μl rehydration buffer (8 M urea, 2% CHAPS, 0.5% IPG buffer, and 0.002% bromophenol blue), and then the lysates were centrifuged at 20,000 g for 10 min. Supernatants were loaded to IEF strips with a program comprising 20 V, 10 hr; 100 V, 1 hr; 500 V, 1 hr; 1,000 V, 1 hr; 2,000 V, 1 hr; 4,000 V, 1 hr; and 8,000 V, 4 hr. Then strips were incubated with SDS equilibration buffer (50 mM Tris-HCl [pH 8.8], 6M urea, 30% glycerol, 2% SDS, 0.001% bromophenol blue) containing 10 mg/ml DTT for 15 min and SDS equilibration buffer containing 2-iodoacetamide for 15 min. Strips were washed with SDS-PAGE buffer, resolved by SDS-PAGE, and analyzed by immunoblotting.
The in vitro deamidation assay was performed as previously described. Briefly, 5 μg of purified MBP-mcGAS or MBP-mcGAS-DDEE mutant on amylose resin was incubated with 0.5 gig of purified FLAG-UL37 or FLAG-UL37C819S at 30° C. for 45 min in deamidation buffer (50 mM Tris-HCl [pH 7.5], 100 mM NaCl, and 5 mM MgCl2). Then the reaction was stopped by adding rehydration buffer and the eluted proteins were analyzed by two-dimensional gel electrophoresis.
cGAMP Reporter Assay
THP-1 Cells were transfected with HT-DNA (2 μg/ml) or infected with HSV-1 virus for 6 h. Cell extracts were prepared by heating at 95° C. for 5 min to denature most proteins, and then the precipitated proteins were removed by centrifugation. The supernatant containing cGAMP was delivered to digitonin-permeabilized (2.5 μg/ml for 30 min) THP1-Lucia cells at 37° C. for 30 min. The cells were cultured for another 20 h before Lucia reporter activity was measured according to the manufacturer's instruction (InvivoGen). Pure cGAMP was diluted and used as standard for the assay.
In Vitro cGAMP Activity Assay
1 μM of MBP-cGAS or mutant proteins was mixed with 100 μM ATP and 100 μM GTP and 10 μCi [α-P32]-ATP in reaction buffer (20 mM Tris-Cl [pH 7.5], 150 mM NaCl, 5 mM MgCl2, 1 mM dithiothreitol [DTT]). After 2 h of incubation at 37° C., 2 μl of reaction solution was spotted onto PEI-Cellulose thin layer chromatography plate (Sigma). Reaction products were resolved with running buffer (1 M (NH4)2SO4, 1.5 M KH2PO4, pH 3.8). The TLC plates were dried and scanned with Phosphoimager (Fuji).
Lentivirus production was carried out in 293T cells as described previously. THP-1 or L929 cells were infected with lentivirus containing UL37 or UL37 C819S mutant. After 36 h, THP-1 cells were selected with puromycin at 1 μg/ml and L929 cells were selected with puromycin at 5 μg/ml.
Purified mcGAS(141-507) was deamidated by purified FLAG-UL37 in vitro. Then the samples were resolved with SDS-PAGE and the mcGAS bands were excised and applied to LC/MS/MS analysis. The analysis of samples was carried out using a Thermo Scientific Q-Exactive hybrid Quadrupole-Orbitrap Mass Spectrometer and a Thermo Dionex UltiMate 3000 RSLCnano System. Peptide mixtures from each sample were loaded onto a peptide trap cartridge at a flow rate of 5 μL/min. The trapped peptides were eluted onto a reversed-phase PicoFrit column (New Objective, Woburn, Mass.) using a linear gradient of acetonitrile (3-36%) in 0.1% formic acid. The elution duration was 120 min at a flow rate of 0.3 μl/min. Eluted peptides from the PicoFrit column were ionized and sprayed into the mass spectrometer, using a Nanospray Flex Ion Source ES071 (Thermo) under the following settings: spray voltage, 2.0 kV, Capillary temperature, 250° C. Other settings were empirically determined. Raw data files were searched against mouse protein sequence database obtained from NCBI website using the Proteome Discoverer 1.4 software (Thermo, San Jose, Calif.) based on the SEQUEST algorithm. Carbamidomethylation (+57.021 Da) of cysteines was fixed modification, and Oxidation and Deamidation Q/N-deamidated (+0.98402 Da) were set as dynamic modifications. The minimum peptide length was specified to be five amino acids. The precursor mass tolerance was set to 15 ppm, whereas fragment mass tolerance was set to 0.05 Da. The maximum false peptide discovery rate was specified as 0.01. The resulting Proteome Discoverer Report contains all assembled proteins with peptides sequences and matched spectrum counts and peak area.
HSV-1-infected mice were sacrificed at 4, 6, 8 days post-infection (dpi) and spleen were collected. Single cell suspension was generated by passing through 40 μM strainer on ice. Red blood cells were removed by adding 5 ml of Pharm Lysis buffer (BD Biosciences). Cells were washed once with cold PBS plus 1% FBS and subjected to tetramer staining.
Tetramer staining was carried out as previously described. Briefly, cells were incubated with anti-CD16/32 antibody for 10 min on ice, followed by staining for 1 h in the dark with tetramers (1:100). Then the cells were stained with anti-CD8 antibody for 20 min on ice. Samples were analyzed by flow cytometry using FACSCalibur and data were analyzed with FlowJo software.
Mouse tissue samples were fixed in 10% (vol/vol) formalin solution (Sigma) overnight. Tissue specimens were dehydrated, embedded in paraffin, and cut into 8-μm sections. Tissue sections were analyzed by H&E and Images were collected with Keyence BZ-X700 microscope.
For immunohistochemistry staining, mouse tissue samples were fixed with 10% formalin solution overnight. Tissue specimens were dehydrated, embedded in paraffin, and cut into 8-μm sections. Tissue sections were analyzed by immunohistochemistry staining with antibodies against UL37, VHS and DAB substrate kit (Vector Laboratories). Images were visualized with Keyence BZ-X700 microscope.
HSV-1-specific antibody detection was carried out as previously described. HSV-1 was purified by ultracentrifuge at 32,000 rpm for 2.5 h and concentrated viral particles were coated to ELISA plates at 4° C. overnight. Plates were then washed five times with PBS-Tween (0.05%) (PBST) and blocked with 1% BSA for 2 h at room temperature. Two-fold dilutions of sera, starting with an initial dilution of 1:10 in PBST were added to the wells and the plates were incubated at RT for 2 h. After washing, rabbit anti-mouse immunoglobulin conjugated to horseradish peroxidase (HRP) diluted at 1:5,000 was added and the plates were incubated for 1 h at room temperature. HSV-1-specific antibody was detected by adding TMB substrate (BD biosciences) and the absorbance was measured at 450 nm. Standard curve was generated by using mouse anti-HSV-1 antiserum. Antibody levels were expressed as arbitrary units against the standard.
THP-1 cells were stimulated with HT-DNA (2 μg/ml) or cGAMP (2 μg/ml) for 16 h or cells were infected with HSV-1 for 16 h. The medium were collected and applied to cytokine measurement. Mice were infected with HSV-1 or HSV-1 UL37 C819S virus (5×107) and the sera were collected 8 hours post-infection.
ELISA kit for human interferon-β (PBL assay science) was used to determine the concentration of human interferon-β. ELISA kits for murine interferon-α (PBL assay science), interferon-β (PBL assay science), CCL5 (R&D systems) and IL-6 (BD Biosciences) were used to determine the concentration of cytokines in the mouse serum according to the manufacturer's instructions.
It is to be understood that while the disclosure has been described in conjunction with the above embodiments, that the foregoing description and examples are intended to illustrate and not limit the scope of the disclosure. Other aspects, advantages and modifications within the scope of the disclosure will be apparent to those skilled in the art to which the disclosure pertains.
The embodiments illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising,” “including,” containing,” etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the disclosure.
Thus, it should be understood that although the present disclosure has been specifically disclosed by specific embodiments and optional features, modification, improvement and variation of the embodiments therein herein disclosed may be resorted to by those skilled in the art, and that such modifications, improvements and variations are considered to be within the scope of this disclosure. The materials, methods, and examples provided here are representative of particular embodiments, are exemplary, and are not intended as limitations on the scope of the disclosure.
The scoped of the disclosure has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the disclosure. This includes the generic description with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that embodiments of the disclosure may also thereby be described in terms of any individual member or subgroup of members of the Markush group.
All publications, patent applications, patents, and other references mentioned herein are expressly incorporated by reference in their entirety, to the same extent as if each were incorporated by reference individually. In case of conflict, the present specification, including definitions, will control.
This application claims the benefit under 35 U.S.C. 119(e) of U.S. Provisional Ser. No. 62/414,592, filed Oct. 28, 2016, the contents of which is hereby incorporated by reference into the present disclosure.
This invention was made with government support under the Grant No. DE021445, awarded by the National Institute for Health. Accordingly, the U.S. Government has certain rights to the invention.
Number | Date | Country | |
---|---|---|---|
62414592 | Oct 2016 | US |