Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cell ; 163(3): 774-774.e1, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496613

ABSTRACT

Restriction factors are cellular proteins that inhibit viruses at different steps of their replication cycle and represent an important first line of defense against viral pathogens. This SnapShot provides an overview of cell-intrinsic antiviral factors, describes their properties, and illustrates the striking variety of antiviral mechanisms as well the sophisticated viral countermeasures. To view this SnapShot, open or download the PDF.


Subject(s)
Cells/virology , Host-Pathogen Interactions , Virus Diseases/metabolism , Animals , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Virus Diseases/immunology , Viruses/metabolism
2.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29976668

ABSTRACT

The host restriction factor tetherin inhibits virion release from infected cells and poses a significant barrier to successful zoonotic transmission of primate lentiviruses to humans. While most simian immunodeficiency viruses (SIV), including the direct precursors of human immunodeficiency virus type 1 (HIV-1) and HIV-2, use their Nef protein to counteract tetherin in their natural hosts, they fail to antagonize the human tetherin ortholog. Pandemic HIV-1 group M and epidemic group O strains overcame this hurdle by adapting their Vpu and Nef proteins, respectively, whereas HIV-2 group A uses its envelope (Env) glycoprotein to counteract human tetherin. Whether or how the remaining eight groups of HIV-2 antagonize this antiviral factor has remained unclear. Here, we show that Nef proteins from diverse groups of HIV-2 do not or only modestly antagonize human tetherin, while their ability to downmodulate CD3 and CD4 is highly conserved. Experiments in transfected cell lines and infected primary cells revealed that not only Env proteins of epidemic HIV-2 group A but also those of a circulating recombinant form (CRF01_AB) and rare groups F and I decrease surface expression of human tetherin and significantly enhance progeny virus release. Intriguingly, we found that many SIVsmm Envs also counteract human as well as smm tetherin. Thus, Env-mediated tetherin antagonism in different groups of HIV-2 presumably stems from a preadaptation of their SIVsmm precursors to humans. In summary, we identified a phenotypic trait of SIVsmm that may have facilitated its successful zoonotic transmission to humans and the emergence of HIV-2.IMPORTANCE HIV-2 groups A to I resulted from nine independent cross-species transmission events of SIVsmm to humans and differ considerably in their prevalence and geographic spread. Thus, detailed characterization of these viruses offers a valuable means to elucidate immune evasion mechanisms and human-specific adaptations determining viral spread. In a systematic comparison of rare and epidemic HIV-2 groups and their simian SIVsmm counterparts, we found that the ability of Nef to downmodulate the primary viral entry receptor CD4 and the T cell receptor CD3 is conserved, while effects on CD28, CD74, and major histocompatibility complex class I surface expression vary considerably. Furthermore, we show that not only the Env proteins of HIV-2 groups A, AB, F, and I but also those of some SIVsmm isolates antagonize human tetherin. This finding helps to explain why SIVsmm has been able to cross the species barrier to humans on at least nine independent occasions.


Subject(s)
Antigens, CD/genetics , Gene Products, nef/genetics , HIV-2/genetics , Simian Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/genetics , Antigens, CD/metabolism , CD3 Complex/genetics , CD4 Antigens/genetics , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HIV-2/metabolism , Host-Pathogen Interactions , Humans , Simian Immunodeficiency Virus/metabolism
3.
J Virol ; 89(23): 12154-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26401043

ABSTRACT

UNLABELLED: Tetherin is an interferon-inducible restriction factor targeting a broad range of enveloped viruses. Its antiviral activity depends on an unusual topology comprising an N-terminal transmembrane domain (TMD) followed by an extracellular coiled-coil region and a C-terminal glycosylphosphatidylinositol (GPI) anchor. One of the two membrane anchors is inserted into assembling virions, while the other remains in the plasma membrane of the infected cell. Thus, tetherin entraps budding viruses by physically bridging viral and cellular membranes. Although tetherin restricts the release of a large variety of diverse human and animal viruses, only mammalian orthologs have been described to date. Here, we examined the evolutionary origin of this protein and demonstrate that tetherin orthologs are also found in fish, reptiles, and birds. Notably, alligator tetherin efficiently blocks the release of retroviral particles. Thus, tetherin emerged early during vertebrate evolution and acquired its antiviral activity before the mammal/reptile divergence. Although there is only limited sequence homology, all orthologs share the typical topology. Two unrelated proteins of the slime mold Dictyostelium discoideum also adopt a tetherin-like configuration with an N-terminal TMD and a C-terminal GPI anchor. However, these proteins showed no evidence for convergent evolution and failed to inhibit virion release. In summary, our findings demonstrate that tetherin emerged at least 450 million years ago and is more widespread than previously anticipated. The early evolution of antiviral activity together with the high topology conservation but low sequence homology suggests that restriction of virus release is the primary function of tetherin. IMPORTANCE: The continuous arms race with viruses has driven the evolution of a variety of cell-intrinsic immunity factors that inhibit different steps of the viral replication cycle. One of these restriction factors, tetherin, inhibits the release of newly formed progeny virions from infected cells. Although tetherin targets a broad range of enveloped viruses, including retro-, filo-, herpes-, and arenaviruses, the evolutionary origin of this restriction factor and its antiviral activity remained obscure. Here, we examined diverse vertebrate genomes for genes encoding cellular proteins that share with tetherin the highly unusual combination of an N-terminal transmembrane domain and a C-terminal glycosylphosphatidylinositol anchor. We show that tetherin orthologs are found in fish, reptiles, and birds and demonstrate that alligator tetherin efficiently inhibits the release of retroviral particles. Our findings identify tetherin as an evolutionarily ancient restriction factor and provide new important insights into the continuous arms race between viruses and their hosts.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Carrier Proteins/genetics , Evolution, Molecular , Microfilament Proteins/genetics , Virus Release/physiology , Animals , Base Sequence , CHO Cells , Cluster Analysis , Computational Biology , Cricetinae , Cricetulus , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA , Species Specificity , Virus Release/genetics
4.
Retrovirology ; 12: 41, 2015 May 16.
Article in English | MEDLINE | ID: mdl-25980612

ABSTRACT

BACKGROUND: Known antiretroviral restriction factors are encoded by genes that are under positive selection pressure, induced during HIV-1 infection, up-regulated by interferons, and/or interact with viral proteins. To identify potential novel restriction factors, we performed genome-wide scans for human genes sharing molecular and evolutionary signatures of known restriction factors and tested the anti-HIV-1 activity of the most promising candidates. RESULTS: Our analyses identified 30 human genes that share characteristics of known restriction factors. Functional analyses of 27 of these candidates showed that over-expression of a strikingly high proportion of them significantly inhibited HIV-1 without causing cytotoxic effects. Five factors (APOL1, APOL6, CD164, TNFRSF10A, TNFRSF10D) suppressed infectious HIV-1 production in transfected 293T cells by >90% and six additional candidates (FCGR3A, CD3E, OAS1, GBP5, SPN, IFI16) achieved this when the virus was lacking intact accessory vpr, vpu and nef genes. Unexpectedly, over-expression of two factors (IL1A, SP110) significantly increased infectious HIV-1 production. Mechanistic studies suggest that the newly identified potential restriction factors act at different steps of the viral replication cycle, including proviral transcription and production of viral proteins. Finally, we confirmed that mRNA expression of most of these candidate restriction factors in primary CD4+ T cells is significantly increased by type I interferons. CONCLUSIONS: A limited number of human genes share multiple characteristics of genes encoding for known restriction factors. Most of them display anti-retroviral activity in transient transfection assays and are expressed in primary CD4+ T cells.


Subject(s)
HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions , Immunity, Innate , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Line , Gene Expression Profiling , Genetic Testing , Humans
5.
PLoS Pathog ; 8(12): e1003093, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23308067

ABSTRACT

HIV-1 groups M and N emerged within the last century following two independent cross-species transmissions of SIVcpz from chimpanzees to humans. In contrast to pandemic group M strains, HIV-1 group N viruses are exceedingly rare, with only about a dozen infections identified, all but one in individuals from Cameroon. Poor adaptation to the human host may be responsible for this limited spread of HIV-1 group N in the human population. Here, we analyzed the function of Vpu proteins from seven group N strains from Cameroon, the place where this zoonosis originally emerged. We found that these N-Vpus acquired four amino acid substitutions (E15A, V19A and IV25/26LL) in their transmembrane domain (TMD) that allow efficient interaction with human tetherin. However, despite these adaptive changes, most N-Vpus still antagonize human tetherin only poorly and fail to down-modulate CD4, the natural killer (NK) cell ligand NTB-A as well as the lipid-antigen presenting protein CD1d. These functional deficiencies were mapped to amino acid changes in the cytoplasmic domain that disrupt putative adaptor protein binding sites and an otherwise highly conserved ßTrCP-binding DSGxxS motif. As a consequence, N-Vpus exhibited aberrant intracellular localization and/or failed to recruit the ubiquitin-ligase complex to induce tetherin degradation. The only exception was the Vpu of a group N strain recently discovered in France, but originally acquired in Togo, which contained intact cytoplasmic motifs and counteracted tetherin as effectively as the Vpus of pandemic HIV-1 M strains. These results indicate that HIV-1 group N Vpu is under strong host-specific selection pressure and that the acquisition of effective tetherin antagonism may lead to the emergence of viral variants with increased transmission fitness.


Subject(s)
Antigens, CD/metabolism , HIV Infections/virology , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Selection, Genetic , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Cells, Cultured , Flow Cytometry , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , HIV Infections/metabolism , HIV-1/pathogenicity , Humans , Molecular Sequence Data , Protein Interaction Domains and Motifs , Sequence Homology, Amino Acid , Virus Release
6.
Retrovirology ; 10: 32, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23514615

ABSTRACT

BACKGROUND: The acquisition of effective Vpu-mediated anti-tetherin activity to promote virion release following transmission of SIVcpzPtt from central chimpanzees (Pan troglodytes troglodytes) to humans distinguishes pandemic HIV-1 group M strains from non-pandemic group N, O and P viruses and may have been a prerequisite for their global spread. Some functional motifs in the cytoplasmic region of HIV-1 M Vpus proposed to be important for anti-tetherin activity are more frequently found in the Vpu proteins of SIVcpzPtt than in those of SIVcpzPts infecting eastern chimpanzees (P. t. schweinfurthii), that have not been detected in humans, and SIVgor from gorillas, which is closely related to HIV-1 O and P. Thus, SIVcpzPtt strains may require fewer adaptive changes in Vpu than SIVcpzPts or SIVgor strains to counteract human tetherin. RESULTS: To examine whether SIVcpzPtt may only need changes in the transmembrane domain (TMD) of Vpu to acquire anti-tetherin activity, whereas SIVcpzPts and SIVgor may also require changes in the cytoplasmic region, we analyzed chimeras between the TMD of an HIV-1 M Vpu and the cytoplasmic domains of SIVcpzPtt (n = 2), SIVcpzPts (n = 2) and SIVgor (n = 2) Vpu proteins. Unexpectedly, all of these chimeras were capable of counteracting human tetherin to enhance virion release, irrespective of the presence or absence of the putative adaptor protein binding sites and the DSGxxS ß-TrCP binding motif reported to be critical for effective anti-tetherin activity of M Vpus. It was also surprising that in three of the six chimeras the gain of anti-tetherin function was associated with a loss of the CD4 degradation activity since this function was conserved among all parental HIV-1, SIVcpz and SIVgor Vpu proteins. CONCLUSIONS: Our results show that changes in the TMD of SIVcpzPtt, SIVcpzPts and SIVgor Vpus are sufficient to render them active against human tetherin. Thus, several previously described domains in the extracellular region of Vpu are not absolutely essential for tetherin antagonism but may be required for other Vpu functions.


Subject(s)
HIV-1/physiology , Human Immunodeficiency Virus Proteins/metabolism , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins/metabolism , Virus Release , Animals , Antigens, CD , Cell Line , GPI-Linked Proteins/antagonists & inhibitors , Gorilla gorilla , HIV-1/genetics , Human Immunodeficiency Virus Proteins/genetics , Humans , Pan troglodytes , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics
7.
Cell Host Microbe ; 19(4): 504-14, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-26996307

ABSTRACT

Guanylate binding proteins (GBPs) are an interferon (IFN)-inducible subfamily of guanosine triphosphatases (GTPases) with well-established activity against intracellular bacteria and parasites. Here we show that GBP5 potently restricts HIV-1 and other retroviruses. GBP5 is expressed in the primary target cells of HIV-1, where it impairs viral infectivity by interfering with the processing and virion incorporation of the viral envelope glycoprotein (Env). GBP5 levels in macrophages determine and inversely correlate with infectious HIV-1 yield over several orders of magnitude, which may explain the high donor variability in macrophage susceptibility to HIV. Antiviral activity requires Golgi localization of GBP5, but not its GTPase activity. Start codon mutations in the accessory vpu gene from macrophage-tropic HIV-1 strains conferred partial resistance to GBP5 inhibition by increasing Env expression. Our results identify GBP5 as an antiviral effector of the IFN response and may explain the increased frequency of defective vpu genes in primary HIV-1 strains.


Subject(s)
GTP-Binding Proteins/immunology , HIV Infections/enzymology , HIV-1/physiology , Interferons/immunology , GTP-Binding Proteins/genetics , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Macrophages/immunology , Macrophages/virology , Protein Transport , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
8.
Virology ; 482: 72-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25827531

ABSTRACT

Heterosexual HIV-1 transmission has been identified as a genetic bottleneck and a single transmitted/founder (T/F) variant with reduced sensitivity to type I interferon initiates productive infection in most cases. We hypothesized that particularly active accessory protein(s) may confer T/F viruses with a selective advantage in establishing HIV infection. Thus, we tested vpu, vif and nef alleles from six T/F and six chronic (CC) viruses in assays for 9 immune evasion activities involving the counteraction of interferon-stimulated genes and modulation of ligands known to activate innate immune cells. All functions were highly conserved with no significant differences between T/F and CC viruses, suggesting that these accessory protein functions are important throughout the course of infection.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , HIV-1/physiology , Human Immunodeficiency Virus Proteins/metabolism , Immune Evasion , Viral Regulatory and Accessory Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Female , HIV Infections/virology , HIV-1/isolation & purification , Humans , Male , Virus Replication
9.
Cell Rep ; 10(4): 586-99, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25620704

ABSTRACT

NF-κB is essential for effective transcription of primate lentiviral genomes and also activates antiviral host genes. Here, we show that the early protein Nef of most primate lentiviruses enhances NF-κB activation. In contrast, the late protein Vpu of HIV-1 and its simian precursors inhibits activation of NF-κB, even in the presence of Nef. Although this effect of Vpu did not correlate with its ability to interact with ß-TrCP, it involved the stabilization of IκB and reduced nuclear translocation of p65. Interestingly, however, Vpu did not affect casein kinase II-mediated phosphorylation of p65. Lack of Vpu was associated with increased NF-κB activation and induction of interferon and interferon-stimulated genes (ISGs) in HIV-1-infected T cells. Thus, HIV-1 and its simian precursors employ Nef to boost NF-κB activation early during the viral life cycle to initiate proviral transcription, while Vpu is used to downmodulate NF-κB-dependent expression of ISGs at later stages.


Subject(s)
Lentiviruses, Primate/metabolism , NF-kappa B/metabolism , Viral Proteins/metabolism , Animals , Gene Expression , HIV-1/genetics , HIV-1/metabolism , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Immunity, Innate/physiology , Lentiviruses, Primate/genetics , Viral Proteins/genetics
10.
Cell Rep ; 11(5): 737-47, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921529

ABSTRACT

CXCL12-CXCR4 signaling controls multiple physiological processes and its dysregulation is associated with cancers and inflammatory diseases. To discover as-yet-unknown endogenous ligands of CXCR4, we screened a blood-derived peptide library for inhibitors of CXCR4-tropic HIV-1 strains. This approach identified a 16 amino acid fragment of serum albumin as an effective and highly specific CXCR4 antagonist. The endogenous peptide, termed EPI-X4, is evolutionarily conserved and generated from the highly abundant albumin precursor by pH-regulated proteases. EPI-X4 forms an unusual lasso-like structure and antagonizes CXCL12-induced tumor cell migration, mobilizes stem cells, and suppresses inflammatory responses in mice. Furthermore, the peptide is abundant in the urine of patients with inflammatory kidney diseases and may serve as a biomarker. Our results identify EPI-X4 as a key regulator of CXCR4 signaling and introduce proteolysis of an abundant precursor protein as an alternative concept for chemokine receptor regulation.


Subject(s)
Peptide Fragments/metabolism , Peptides/metabolism , Receptors, CXCR4/antagonists & inhibitors , Serum Albumin/metabolism , Amino Acid Sequence , Animals , Biomarkers/urine , Cell Line , Cell Movement/drug effects , HEK293 Cells , HIV-1/physiology , Half-Life , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Library , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Protein Structure, Tertiary , Receptors, CXCR4/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Sequence Alignment , Serum Albumin/chemistry , Serum Albumin/pharmacology , Signal Transduction/drug effects , Virus Internalization/drug effects
11.
Sci Transl Med ; 6(262): 262ra157, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25391483

ABSTRACT

Topically applied microbicides potently inhibit HIV in vitro but have largely failed to exert protective effects in clinical trials. One possible reason for this discrepancy is that the preclinical testing of microbicides does not faithfully reflect the conditions of HIV sexual transmission. We report that candidate microbicides that target HIV components show greatly reduced antiviral efficacy in the presence of semen, the main vector for HIV transmission. This diminished antiviral activity was dependent on the ability of amyloid fibrils in semen to enhance the infectivity of HIV. Thus, the anti-HIV efficacy of microbicides determined in the absence of semen greatly underestimated the drug concentrations needed to block semen-exposed virus. One notable exception was maraviroc. This HIV entry inhibitor targets the host cell CCR5 co-receptor and was highly active against both untreated and semen-exposed HIV. These data help to explain why microbicides have failed to protect against HIV in clinical trials and suggest that antiviral compounds targeting host factors hold promise for further development. These findings also suggest that the in vitro efficacy of candidate microbicides should be determined in the presence of semen to identify the best candidates for the prevention of HIV sexual transmission.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-Infective Agents/pharmacology , HIV Infections/prevention & control , Semen/chemistry , Semen/virology , Cell Line , Cell Line, Tumor , Cell Survival , Dendrimers/pharmacology , Female , HIV Infections/drug therapy , HIV-1/pathogenicity , Humans , Inhibitory Concentration 50 , Polylysine/pharmacology
12.
Cell Host Microbe ; 16(5): 639-50, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25525794

ABSTRACT

Most simian immunodeficiency viruses use their Nef protein to antagonize the host restriction factor tetherin. A deletion in human tetherin confers Nef resistance, representing a hurdle to successful zoonotic transmission. HIV-1 group M evolved to utilize the viral protein U (Vpu) to counteract tetherin. Although HIV-1 group O has spread epidemically in humans, it has not evolved a Vpu-based tetherin antagonism. Here we show that HIV-1 group O Nef targets a region adjacent to this deletion to inhibit transport of human tetherin to the cell surface, enhances virion release, and increases viral resistance to inhibition by interferon-α. The Nef protein of the inferred common ancestor of group O viruses is also active against human tetherin. Thus, Nef-mediated antagonism of human tetherin evolved prior to the spread of HIV-1 group O and likely facilitated secondary virus transmission. Our results may explain the epidemic spread of HIV-1 group O.


Subject(s)
Antigens, CD/genetics , HIV-1/pathogenicity , nef Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line, Tumor , Endocytosis , Evolution, Molecular , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , HIV-1/classification , Humans , Molecular Sequence Data , Protein Conformation , Sequence Analysis , Sequence Deletion , Virion/genetics , Virion/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
13.
Antiviral Res ; 87(2): 272-5, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20488208

ABSTRACT

The NS3/4A serine protease of the hepatitis C virus (HCV) is one of the most attractive targets for specific antiviral agents. However, mutations conferring resistance may decrease the efficacy of these drugs. Although the level of resistance associated with specific mutations differs between different compounds, substitutions R155K and A156T reduce susceptibility to all protease inhibitors published so far. Interestingly, variants harboring the resistant mutation R155K were also detected as the predominant quasispecies in some treatment-naïve patients. Of note, key positions for resistance overlap with the HLA-A*68-restricted epitope HAVGIFRAAV(1175-1184). The aim of our study was to analyze the impact of protease inhibitor resistance mutations on the replication level and the antiviral CD8 T cell response against this HCV epitope. Our findings suggest that the R155K variant is associated with a relatively high replication level and with a substantial loss of cross-recognition by specific CD8 T cells targeting the epitope HAVGIFRAAV(1175-1184), providing a possible explanation for its existence in the absence of drug selection pressure.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/genetics , Drug Resistance, Viral , HLA-A Antigens/immunology , Hepacivirus/immunology , Mutation, Missense , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Amino Acid Substitution/genetics , Antiviral Agents/pharmacology , Carbamates/pharmacology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Hepacivirus/growth & development , Humans , Intracellular Signaling Peptides and Proteins , Macrocyclic Compounds/pharmacology , Quinolines/pharmacology , Thiazoles/pharmacology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL