Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Adv Exp Med Biol ; 1366: 65-85, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35412135

RESUMO

The discovery of the G-protein coupled-receptor (GPCR) CXCR4 as a major coreceptor of HIV-1 entry about three decades ago explained why the chemokine SDF-1/CXCL12 inhibits specific viral strains. The knowledge that RANTES, MlP-1α, and MlP-1ß specifically inhibit other primary HIV-1 strains allowed the rapid discovery of CCR5 as second major viral coreceptor and explained why individuals with deletions in CCR5 are protected against sexual HIV-1 transmission. Here, we provide an update on endogenous ligands of GPCRs that act as endogenous inhibitors of HIV-1, HIV-2, and simian immunodeficiency virus (SIV) entry. In addition, we summarize the development of optimized derivatives of endogenous GPCR ligands and their perspectives as antiviral agents and beyond. Finally, we provide examples for other endogenous peptides that may contribute to our innate immune defense against HIV-1 and other viral pathogens and offer prospects for preventive or therapeutic development.


Assuntos
Inibidores da Fusão de HIV , Infecções por HIV , HIV-1 , Animais , Inibidores da Fusão de HIV/farmacologia , Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1/metabolismo , HIV-1/fisiologia , HIV-2/metabolismo , HIV-2/fisiologia , Humanos , Ligantes , Peptídeos/uso terapêutico , Receptores CCR5 , Receptores Acoplados a Proteínas G/uso terapêutico , Transdução de Sinais , Vírus da Imunodeficiência Símia
2.
J Virol ; 95(23): e0117021, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34523960

RESUMO

Vif is a lentiviral accessory protein that counteracts the antiviral activity of cellular APOBEC3 (A3) cytidine deaminases in infected cells. The exact contribution of each member of the A3 family for the restriction of HIV-2 is still unclear. Thus, the aim of this work was to identify the A3s with anti-HIV-2 activity and compare their restriction potential for HIV-2 and HIV-1. We found that A3G is a strong restriction factor of both types of viruses and A3C restricts neither HIV-1 nor HIV-2. Importantly, A3B exhibited potent antiviral activity against HIV-2, but its effect was negligible against HIV-1. Whereas A3B is packaged with similar efficiency into both viruses in the absence of Vif, HIV-2 and HIV-1 differ in their sensitivity to A3B. HIV-2 Vif targets A3B by reducing its cellular levels and inhibiting its packaging into virions, whereas HIV-1 Vif did not evolve to antagonize A3B. Our observations support the hypothesis that during wild-type HIV-1 and HIV-2 infections, both viruses are able to replicate in host cells expressing A3B but using different mechanisms, probably resulting from a Vif functional adaptation over evolutionary time. Our findings provide new insights into the differences between Vif protein and their cellular partners in the two human viruses. Of note, A3B is highly expressed in some cancer cells and may cause deamination-induced mutations in these cancers. Thus, A3B may represent an important therapeutic target. As such, the ability of HIV-2 Vif to induce A3B degradation could be an effective tool for cancer therapy. IMPORTANCE Primate lentiviruses encode a series of accessory genes that facilitate virus adaptation to its host. Among those, the vif-encoded protein functions primarily by targeting the APOBEC3 (A3) family of cytidine deaminases. All lentiviral Vif proteins have the ability to antagonize A3G; however, antagonizing other members of the A3 family is variable. Here, we report that HIV-2 Vif, unlike HIV-1 Vif, can induce degradation of A3B. Consequently, HIV-2 Vif but not HIV-1 Vif can inhibit the packaging of A3B. Interestingly, while A3B is packaged efficiently into the core of both HIV-1 and HIV-2 virions in the absence of Vif, it only affects the infectivity of HIV-2 particles. Thus, HIV-1 and HIV-2 have evolved two distinct mechanisms to antagonize the antiviral activity of A3B. Aside from its antiviral activity, A3B has been associated with mutations in some cancers. Degradation of A3B by HIV-2 Vif may be useful for cancer therapies.


Assuntos
Citidina Desaminase/metabolismo , Produtos do Gene vif/metabolismo , HIV-1/metabolismo , HIV-2/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Animais , Citidina Desaminase/genética , Células HEK293 , Infecções por HIV , Humanos , Antígenos de Histocompatibilidade Menor/genética , Receptor EphB2
3.
Nat Commun ; 12(1): 3691, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34140527

RESUMO

The HIV-1 accessory proteins Vif, Vpu, and Nef can promote infection by overcoming the inhibitory effects of the host cell restriction factors APOBEC3G, Tetherin, and SERINC5, respectively. However, how the HIV-1 accessory protein Vpr enhances infection in macrophages but not in CD4+ T cells remains elusive. Here, we report that Vpr counteracts lysosomal-associated transmembrane protein 5 (LAPTM5), a potent inhibitor of HIV-1 particle infectivity, to enhance HIV-1 infection in macrophages. LAPTM5 transports HIV-1 envelope glycoproteins to lysosomes for degradation, thereby inhibiting virion infectivity. Vpr counteracts the restrictive effects of LAPTM5 by triggering its degradation via DCAF1. In the absence of Vpr, the silencing of LAPTM5 precisely phenocopied the effect of Vpr on HIV-1 infection. In contrast, Vpr did not enhance HIV-1 infection in the absence of LAPTM5. Moreover, LAPTM5 was highly expressed in macrophages but not in CD4+ T lymphocytes. Re-expressing LAPTM5 reconstituted the Vpr-dependent promotion of HIV-1 infection in primary CD4+ T cells, as observed in macrophages. Herein, we demonstrate the molecular mechanism used by Vpr to overcome LAPTM5 restriction in macrophages, providing a potential strategy for anti-HIV/AIDS therapeutics.


Assuntos
Infecções por HIV/metabolismo , HIV-1/metabolismo , Interações entre Hospedeiro e Microrganismos , Macrófagos/metabolismo , Macrófagos/virologia , Proteínas de Membrana/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Inativação Gênica , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/genética , HIV-1/patogenicidade , HIV-2/metabolismo , HIV-2/patogenicidade , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Lisossomos/metabolismo , Proteínas de Membrana/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estabilidade Proteica , Vírus da Imunodeficiência Símia/metabolismo , Vírus da Imunodeficiência Símia/patogenicidade , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima , Vírion/metabolismo
4.
Virology ; 554: 17-27, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33333348

RESUMO

The family of human APOBEC3 (A3) restriction factors is formed by seven different proteins, A3A-D and A3F-H. Among these A3s, A3B harbors strong restriction activity against several retroviruses, such as SIV, and MLV. How lentiviruses and other retroviruses, prevalent in many primate species, counteract A3B is poorly understood. In this study, we found that A3B strongly inhibited SIVmac and HIV-2 infectivity, which was antagonized by their Vif proteins. Both SIVmac and HIV-2 Vifs diminished the protein level of A3B in viral producer cells, and hindered A3B incorporation into viral particles. We observed that HIV-2 Vif binds A3B and induces its degradation by assembly of an A3-Vif-CUL5-ElonginB/C E3-ligase complex. A3B and HIV-2 Vif localize and interact in the nucleus. In addition, we also found that the accessory protein Bet of prototype foamy virus (PFV) significantly antagonized the anti-SIVmac activity of A3B. Like Vif, Bet prevented the incorporation of A3B into viral particles. However, in contrast to Vif Bet did not induce the degradation of A3B. Rather, Bet binds A3B to block formation of high molecular weight A3B complexes and induces A3B cytoplasmic trapping. In summary, these findings indicate that A3B is recognized by diverse retroviruses and counteracted by virus-specific pathways that could be targeted to inhibit A3B mutating activity in cancers.


Assuntos
Citidina Desaminase/antagonistas & inibidores , Citidina Desaminase/metabolismo , HIV-2/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Proteínas dos Retroviridae/metabolismo , Spumavirus/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Elonguina/genética , Elonguina/metabolismo , Produtos do Gene vif/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Vírus da Imunodeficiência Símia/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Vírion/metabolismo
5.
Brief Bioinform ; 22(4)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-33253374

RESUMO

INTRODUCTION: Most of the typical chemokine receptors (CKRs) have been identified as coreceptors for a variety of human and simian immunodeficiency viruses (HIVs and SIVs). This study evaluated CCRL2 to examine if it was an HIV/SIV coreceptor. METHODS: The Human glioma cell line, NP-2, is normally resistant to infection by HIV and SIV. The cell was transduced with amplified cluster of differentiation 4 (CD4) as a receptor and CCR5, CXCR4 and CCRL2 as coreceptor candidates to produce NP-2/CD4/coreceptor cells (). The cells were infected with multiplicity of infection (MOI) 1.0. Infected cells were detected by indirect immunofluorescence assay (IFA). Multinucleated giant cells (MGC) in syncytia were quantified by Giemsa staining. Proviral DNA was detected by polymerase chain reaction (PCR), and reverse transcriptase (RT) activity was measured. RESULTS: IFA detected viral antigens of the primary isolates, HIV-1HAN2 and HIV-2MIR in infected NP-2/CD4/CCRL2 cells, indicated CCRL2 as a functional coreceptor. IFA results were confirmed by the detection of proviral DNA and measurement of RT-activity in the spent cell supernatants. Additionally, MGC was detected in HIV-2MIR-infected NP-2/CD4/CCCRL2 cells. HIV-2MIR were found more potent users of CCRL2 than HIV-1HAN2. Moreover, GWAS studies, gene ontology and cell signaling pathways of the HIV-associated genes show interaction of CCRL2 with HIV/SIV envelope protein. CONCLUSIONS: In vitro experiments showed CCRL2 to function as a newly identified coreceptor for primary HIV-2 isolates conveniently. The findings contribute additional insights into HIV/SIV transmission and pathogenesis. However, its in vivo relevance still needs to be evaluated. Confirming in vivo relevance, ligands of CCRL2 can be investigated as potential targets for HIV entry-inhibitor drugs.


Assuntos
Infecções por HIV/metabolismo , HIV-2/metabolismo , Receptores CCR/metabolismo , Infecções por HIV/genética , HIV-1/genética , HIV-1/metabolismo , HIV-2/genética , Humanos , Células Jurkat , Receptores CCR/genética
6.
Int J Mol Sci ; 21(18)2020 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-32971935

RESUMO

During the progression of HIV-1 infection, macrophage tropic HIV-1 that use the CCR5 co-receptor undergoes a change in co-receptor use to CXCR4 that is predominately T cell tropic. This change in co-receptor preference makes the virus able to infect T cells. HIV-2 is known to infect MDMs and T cells and is dual tropic. The aim of this study was to elucidate the differential expression profiles of host miRNAs and their role in cells infected with HIV-1/HIV-2. To achieve this goal, a comparative global miRNA expression profile was determined in human PBMCs and MDMs infected with HIV-1/HIV-2. Differentially expressed miRNAs were identified in HIV-1/HIV-2 infected PBMCs and MDMs using the next-generation sequencing (NGS) technique. A comparative global miRNA expression profile in infected MDMs and PBMCs with HIV-1 and HIV-2 identified differential expression of several host miRNAs. These differentially expressed miRNAs are likely to be involved in many signaling pathways, like the p53 signaling pathway, PI3K-Akt signaling pathways, MAPK signaling pathways, FoxO signaling pathway, and viral carcinogenesis. Thus, a comparative study of the differential expression of host miRNAs in MDMs and T cell in response to HIV-1 and HIV-2 infection will help us to identify unique biomarkers that can differentiate HIV-1 and HIV-2 infection.


Assuntos
Infecções por HIV/metabolismo , HIV-1/metabolismo , HIV-2/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , MicroRNAs/biossíntese , Monócitos/metabolismo , Transcriptoma , Infecções por HIV/patologia , Humanos , Macrófagos/patologia , Macrófagos/virologia , Monócitos/patologia , Monócitos/virologia
7.
J Biol Chem ; 295(2): 657-666, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31806704

RESUMO

Unlike activated CD4+ T cells, nondividing macrophages have an extremely small dNTP pool, which restricts HIV-1 reverse transcription. However, rNTPs are equally abundant in both of these cell types and reach much higher concentrations than dNTPs. The greater difference in concentration between dNTPs and rNTPs in macrophages results in frequent misincorporation of noncanonical rNTPs during HIV-1 reverse transcription. Here, we tested whether the highly abundant SAM domain- and HD domain-containing protein 1 (SAMHD1) deoxynucleoside triphosphorylase in macrophages is responsible for frequent rNTP incorporation during HIV-1 reverse transcription. We also assessed whether Vpx (viral protein X), an accessory protein of HIV-2 and some simian immunodeficiency virus strains that targets SAMHD1 for proteolytic degradation, can counteract the rNTP incorporation. Results from biochemical simulation of HIV-1 reverse transcriptase-mediated DNA synthesis confirmed that rNTP incorporation is reduced under Vpx-mediated dNTP elevation. Using HIV-1 vector, we further demonstrated that dNTP pool elevation by Vpx or deoxynucleosides in human primary monocyte-derived macrophages reduces noncanonical rNTP incorporation during HIV-1 reverse transcription, an outcome similarly observed with the infectious HIV-1 89.6 strain. Furthermore, the simian immunodeficiency virus mac239 strain, encoding Vpx, displayed a much lower level of rNTP incorporation than its ΔVpx mutant in macrophages. Finally, the amount of rNMPs incorporated in HIV-1 proviral DNAs remained unchanged for ∼2 weeks in macrophages. These findings suggest that noncanonical rNTP incorporation is regulated by SAMHD1 in macrophages, whereas rNMPs incorporated in HIV-1 proviral DNA remain unrepaired. This suggests a potential long-term DNA damage impact of SAMHD1-mediated rNTP incorporation in macrophages.


Assuntos
Infecções por HIV/metabolismo , HIV/metabolismo , Macrófagos/virologia , Transcrição Reversa , Ribonucleotídeos/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Células Cultivadas , Desoxirribonucleotídeos/genética , Desoxirribonucleotídeos/metabolismo , HIV/genética , Transcriptase Reversa do HIV/metabolismo , HIV-1/genética , HIV-1/metabolismo , HIV-2/genética , HIV-2/metabolismo , Humanos , Células Jurkat , Macrófagos/metabolismo , Mutagênese , Ribonucleotídeos/genética , Proteína 1 com Domínio SAM e Domínio HD/metabolismo
8.
Nat Microbiol ; 3(8): 891-897, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29891865

RESUMO

To evade host immune defences, human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2) have evolved auxiliary proteins that target cell restriction factors. Viral protein X (Vpx) from the HIV-2/SIVsmm lineage enhances viral infection by antagonizing SAMHD1 (refs 1,2), but this antagonism is not sufficient to explain all Vpx phenotypes. Here, through a proteomic screen, we identified another Vpx target-HUSH (TASOR, MPP8 and periphilin)-a complex involved in position-effect variegation3. HUSH downregulation by Vpx is observed in primary cells and HIV-2-infected cells. Vpx binds HUSH and induces its proteasomal degradation through the recruitment of the DCAF1 ubiquitin ligase adaptor, independently from SAMHD1 antagonism. As a consequence, Vpx is able to reactivate HIV latent proviruses, unlike Vpx mutants, which are unable to induce HUSH degradation. Although antagonism of human HUSH is not conserved among all lentiviral lineages including HIV-1, it is a feature of viral protein R (Vpr) from simian immunodeficiency viruses (SIVs) of African green monkeys and from the divergent SIV of l'Hoest's monkey, arguing in favour of an ancient lentiviral species-specific vpx/vpr gene function. Altogether, our results suggest the HUSH complex as a restriction factor, active in primary CD4+ T cells and counteracted by Vpx, therefore providing a molecular link between intrinsic immunity and epigenetic control.


Assuntos
Antígenos de Neoplasias/metabolismo , Lentivirus de Primatas/fisiologia , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteômica/métodos , Proteínas Virais Reguladoras e Acessórias/metabolismo , Linhagem Celular , Regulação para Baixo , Regulação da Expressão Gênica , Células HEK293 , HIV-2/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Células Jurkat , Lentivirus de Primatas/metabolismo , Provírus/metabolismo , Vírus da Imunodeficiência Símia/metabolismo , Células THP-1
9.
J Virol ; 92(14)2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29743354

RESUMO

Human immunodeficiency virus type 1 (HIV-1) and HIV-2 share a striking genomic resemblance; however, variability in the genetic sequence accounts for the presence of unique accessory genes, such as the viral protein X (vpx) gene in HIV-2. Dual infection with both viruses has long been described in the literature, yet the molecular mechanism of how dually infected patients tend to do better than those who are monoinfected with HIV-1 has not yet been explored. We hypothesized that in addition to extracellular mechanisms, an HIV-2 accessory gene is the culprit, and interference at the viral accessory/regulatory protein level is perhaps responsible for the attenuated pathogenicity of HIV-1 observed in dually infected patients. Following simulation of dual infection in cell culture experiments, we found that pretransduction of cells with HIV-2 significantly protects against HIV-1 transduction. Importantly, we have found that this dampening of the infectivity of HIV-1 was a result of interviral interference carried out by viral protein X of HIV-2, resulting in a severe hindrance to the replication dynamics of HIV-1, influencing both its early and late phases of the viral life cycle. Our findings shed light on potential intracellular interactions between the two viruses and broaden our understanding of the observed clinical spectrum in dually infected patients, highlighting HIV-2 Vpx as a potential candidate worth exploring in the fight against HIV-1.IMPORTANCE Dual infection with human immunodeficiency virus types 1 and 2 is relatively common in areas of endemicity. For as-yet-unclarified reasons, patients who are dually infected were shown to have lower viral loads and generally a lower rate of progression to AIDS than those who are monoinfected. We aimed to explore dual infection in cell culture, to elucidate possible mechanisms by which HIV-2 may be able to exert such an effect. Our results indicate that on the cellular level, pretransduction of cells with HIV-2 significantly protects against HIV-1 transduction, which was found to be a result of interviral interference carried out by viral protein X of HIV-2. These findings broaden our knowledge of interviral interactions on the cellular level and may provide an explanation for the decreased pathogenicity of HIV-1 in dually infected patients, highlighting HIV-2 Vpx as a potential candidate worth exploring in the fight against HIV.


Assuntos
Transformação Celular Viral , Infecções por HIV/prevenção & controle , HIV-1/patogenicidade , HIV-2/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral , Células HEK293 , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Humanos , Proteínas Virais Reguladoras e Acessórias/genética
10.
Front Immunol ; 9: 2978, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619335

RESUMO

Viruses and hosts are situated in a molecular arms race. To avoid morbidity and mortality, hosts evolved antiviral restriction factors. These restriction factors exert selection pressure on the viruses and drive viral evolution toward increasingly efficient immune antagonists. Numerous viruses exploit cellular DNA damage-binding protein 1 (DDB1)-containing Cullin RocA ubiquitin ligases (CRLs) to induce the ubiquitination and subsequent proteasomal degradation of antiviral factors expressed by their hosts. To establish a comprehensive understanding of the underlying protein interaction networks, we performed immuno-affinity precipitations for a panel of DDB1-interacting proteins derived from viruses such as mouse cytomegalovirus (MCMV, Murid herpesvirus [MuHV] 1), rat cytomegalovirus Maastricht MuHV2, rat cytomegalovirus English MuHV8, human cytomegalovirus (HCMV), hepatitis B virus (HBV), and human immunodeficiency virus (HIV). Cellular interaction partners were identified and quantified by mass spectrometry (MS) and validated by classical biochemistry. The comparative approach enabled us to separate unspecific interactions from specific binding partners and revealed remarkable differences in the strength of interaction with DDB1. Our analysis confirmed several previously described interactions like the interaction of the MCMV-encoded interferon antagonist pM27 with STAT2. We extended known interactions to paralogous proteins like the interaction of the HBV-encoded HBx with different Spindlin proteins and documented interactions for the first time, which explain functional data like the interaction of the HIV-2-encoded Vpr with Bax. Additionally, several novel interactions were identified, such as the association of the HIV-2-encoded Vpx with the transcription factor RelA (also called p65). For the latter interaction, we documented a functional relevance in antagonizing NF-κB-driven gene expression. The mutation of the DDB1 binding interface of Vpx significantly impaired NF-κB inhibition, indicating that Vpx counteracts NF-κB signaling by a DDB1- and CRL-dependent mechanism. In summary, our findings improve the understanding of how viral pathogens hijack cellular DDB1 and CRLs to ensure efficient replication despite the expression of host restriction factors.


Assuntos
HIV-2/imunologia , Ligação Proteica/imunologia , Fator de Transcrição RelA/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Viroses/imunologia , Animais , Citomegalovirus/imunologia , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Fibroblastos , Regulação da Expressão Gênica/imunologia , Células HEK293 , HIV-2/genética , HIV-2/metabolismo , Vírus da Hepatite B/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoprecipitação/métodos , Espectrometria de Massas/métodos , Camundongos , Muromegalovirus/imunologia , Células NIH 3T3 , Cultura Primária de Células , Mapeamento de Interação de Proteínas/métodos , Fator de Transcrição RelA/imunologia , Fator de Transcrição RelA/metabolismo , Ubiquitina-Proteína Ligases/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/imunologia , Viroses/virologia
11.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795437

RESUMO

Human immunodeficiency virus type 2 (HIV-2) has already spread to different regions worldwide, and currently about 1 to 2 million people have been infected, calling for new antiviral agents that are effective on both HIV-1 and HIV-2 isolates. T20 (enfuvirtide), a 36-mer peptide derived from the C-terminal heptad repeat region (CHR) of gp41, is the only clinically approved HIV-1 fusion inhibitor, but it easily induces drug resistance and is not active on HIV-2. In this study, we first demonstrated that the M-T hook structure was also vital to enhancing the binding stability and inhibitory activity of diverse CHR-based peptide inhibitors. We then designed a novel short peptide (23-mer), termed 2P23, by introducing the M-T hook structure, HIV-2 sequences, and salt bridge-forming residues. Promisingly, 2P23 was a highly stable helical peptide with high binding to the surrogate targets derived from HIV-1, HIV-2, and simian immunodeficiency virus (SIV). Consistent with this, 2P23 exhibited potent activity in inhibiting diverse subtypes of HIV-1 isolates, T20-resistant HIV-1 mutants, and a panel of primary HIV-2 isolates, HIV-2 mutants, and SIV isolates. Therefore, we conclude that 2P23 has high potential to be further developed for clinical use, and it is also an ideal tool for exploring the mechanisms of HIV-1/2- and SIV-mediated membrane fusion. IMPORTANCE: The peptide drug T20 is the only approved HIV-1 fusion inhibitor, but it is not active on HIV-2 isolates, which have currently infected 1 to 2 million people and continue to spread worldwide. Recent studies have demonstrated that the M-T hook structure can greatly enhance the binding and antiviral activities of gp41 CHR-derived inhibitors, especially for short peptides that are otherwise inactive. By combining the hook structure, HIV-2 sequence, and salt bridge-based strategies, the short peptide 2P23 has been successfully designed. 2P23 exhibits prominent advantages over many other peptide fusion inhibitors, including its potent and broad activity on HIV-1, HIV-2, and even SIV isolates, its stability as a helical, oligomeric peptide, and its high binding to diverse targets. The small size of 2P23 would benefit its synthesis and significantly reduce production cost. Therefore, 2P23 is an ideal candidate for further development, and it also provides a novel tool for studying HIV-1/2- and SIV-mediated cell fusion.


Assuntos
Proteína gp41 do Envelope de HIV/antagonistas & inibidores , Inibidores da Fusão de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-2/efeitos dos fármacos , Peptídeos/farmacologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Sítios de Ligação , Desenho de Fármacos , Farmacorresistência Viral/efeitos dos fármacos , Enfuvirtida , Proteína gp41 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/farmacologia , Inibidores da Fusão de HIV/síntese química , HIV-1/química , HIV-1/metabolismo , HIV-2/química , HIV-2/metabolismo , Humanos , Fragmentos de Peptídeos/farmacologia , Peptídeos/síntese química , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Vírus da Imunodeficiência Símia/química , Vírus da Imunodeficiência Símia/metabolismo , Relação Estrutura-Atividade , Internalização do Vírus/efeitos dos fármacos
12.
J Virol ; 90(22): 10065-10073, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27535053

RESUMO

Binding of human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) gp120 exterior envelope glycoprotein to CD4 triggers conformational changes in gp120 that promote its interaction with one of the chemokine receptors, usually CCR5, ultimately leading to gp41-mediated virus-cell membrane fusion and entry. We previously described that topological layers (layer 1, layer 2, and layer 3) in the gp120 inner domain contribute to gp120-trimer association in the unliganded state but also help secure CD4 binding. Relative to layer 1 of HIV-1 gp120, the SIVmac239 gp120 layer 1 plays a more prominent role in maintaining gp120-trimer association but is minimally involved in promoting CD4 binding, which could be explained by the existence of a well-conserved tryptophan at position 375 (Trp 375) in HIV-2/SIVsmm. In this study, we investigated the role of SIV layer 3 in viral entry, cell-to-cell fusion, and CD4 binding. We observed that a network of interactions involving some residues of the ß8-α5 region in SIVmac239 layer 3 may contribute to CD4 binding by helping shape the nearby Phe 43 cavity, which directly contacts CD4. In summary, our results suggest that layer 3 in SIV has a greater impact on CD4 binding than in HIV-1. This work defines lineage-specific differences in layer 3 from HIV-1 and that from SIV. IMPORTANCE: CD4-induced conformational changes in the gp120 inner domain involve rearrangements between three topological layers. While the role of layers 1 to 3 for HIV-1 and layers 1 and 2 for SIV on gp120 transition to the CD4-bound conformation has been reported, the role of SIV layer 3 remains unknown. Here we report that SIV layer 3 has a greater impact on CD4 binding than does layer 3 in HIV-1 gp120. This work defines lineage-specific differences in layer 3 from HIV-1 and SIV.


Assuntos
Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Vírus da Imunodeficiência Símia/metabolismo , Proteínas do Envelope Viral/metabolismo , Antígenos CD4/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Proteína gp41 do Envelope de HIV/metabolismo , Infecções por HIV/virologia , HIV-2/metabolismo , Células HeLa , Humanos , Ligação Proteica/fisiologia , Conformação Proteica , Receptores CCR5/metabolismo , Internalização do Vírus
13.
PLoS One ; 11(7): e0158941, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27392064

RESUMO

Although human immunodeficiency virus (HIV) types 1 and 2 are closely related lentiviruses with similar replication cycles, HIV-2 infection is associated with slower progression to AIDS, a higher proportion of long term non-progressors, and lower rates of transmission than HIV-1, likely as a consequence of a lower viral load during HIV-2 infection. A mechanistic explanation for the differential viral load remains unclear but knowledge of differences in particle production between HIV-1 and HIV-2 may help to shed light on this issue. In contrast to HIV-1, little is known about the assembly of HIV-2 particles, and the trafficking of HIV-2 Gag, the structural component of the virus, within cells. We have established that HIV-2 Gag accumulates in intracellular CD63 positive compartments, from which it may be delivered or recycled to the cell surface, or degraded. HIV-2 particle release was dependent on the adaptor protein complex AP-3 and the newly identified AP-5 complex, but much less so on AP-1. In contrast, HIV-1 particle release required AP-1 and AP-3, but not AP-5. AP-2, an essential component of clathrin-mediated endocytosis, which was previously shown to be inhibitory to HIV-1 particle release, had no effect on HIV-2. The differential requirement for adaptor protein complexes confirmed that HIV-1 and HIV-2 Gag have distinct cellular trafficking pathways, and that HIV-2 particles may be more susceptible to degradation prior to release.


Assuntos
Complexo 3 de Proteínas Adaptadoras/metabolismo , Subunidades beta do Complexo de Proteínas Adaptadoras/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , HIV-2/metabolismo , Vírion/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Complexo 3 de Proteínas Adaptadoras/genética , Subunidades beta do Complexo de Proteínas Adaptadoras/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , HIV-1/genética , HIV-1/metabolismo , HIV-2/genética , Células HeLa , Humanos , Transporte Proteico , Tetraspanina 30/genética , Tetraspanina 30/metabolismo , Vírion/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
14.
FEBS Lett ; 589(13): 1505-14, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-25936766

RESUMO

In this study, we performed a mutational analysis to determine whether the mechanism by which HIV-2 Vpx confers the capacity for infectivity and viral replication in macrophages is solely dependent on its ability to degrade the host antiviral factor SAMHD1. Contrary to expectations, we demonstrated that P(109) in the C-terminal poly-proline motif of HIV-2 Vpx has two unique roles: to facilitate the specific degradation of SAMHD1 in macrophages, and to facilitate multimerization of Vpx, therefore preventing SAMHD1 degradation in the presence of high levels of Vpx.


Assuntos
HIV-2/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Prolina/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular Tumoral , Células HEK293 , HIV-2/genética , Humanos , Immunoblotting , Dados de Sequência Molecular , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Prolina/genética , Proteólise , Proteína 1 com Domínio SAM e Domínio HD , Proteínas Virais Reguladoras e Acessórias/genética
15.
J Biol Chem ; 289(13): 9146-57, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24532789

RESUMO

Interferon regulatory factor (IRF) family members have been implicated as critical transcription factors that function in immune responses, hematopoietic differentiation, and cell growth regulation. Activation of IRF5 results in the production of pro-inflammatory cytokines such as TNFα, IL6, and IL12, as well as type I interferons. In this study, we demonstrate that HIV-2 Vpx interacts with IRF5, and Vpx inhibits IRF5-mediated transactivation. Expression of Vpx in THP-1 cells reduced mRNA levels and protein production of Toll-like receptor-dependent IL6, IL12p40, and TNFα induced by lipopolysaccharide, R848, and ODN2216. Chromatin immunoprecipitation assays show that Vpx expression results in decreased promoter binding activity of IRF5. This study provides new insights into mechanisms employed by HIV-2 to counteract innate immune defenses against viral infection.


Assuntos
HIV-2/fisiologia , Fatores Reguladores de Interferon/antagonistas & inibidores , Fatores Reguladores de Interferon/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas de Transporte/metabolismo , Citocinas/biossíntese , DNA/metabolismo , Células HEK293 , HIV-2/metabolismo , Humanos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases , Ativação Transcricional , Ubiquitina-Proteína Ligases , Replicação Viral
16.
J Biol Chem ; 287(49): 41210-7, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23076149

RESUMO

SAMHD1 is a newly identified restriction factor that targets lentiviruses in myeloid cells and is countered by the SIV(SM)/HIV-2 Vpx protein. By analyzing a large panel of Vpx mutants, we identify several residues throughout the 3-helix bundle predicted for Vpx that impair both its functionality and its ability to degrade SAMHD1. We determine that SAMHD1 is a strictly non-shuttling nuclear protein and that as expected WT Vpx localizes with it in the nucleus. However, we also identify a functional Vpx mutant with predominant cytoplasmic distribution that colocalizes with SAMHD1 in this location, suggesting that Vpx may also retain SAMHD1 in the cell cytoplasm, prior to its entry into the nucleus. Several mutations in Vpx were shown to affect the stability of Vpx, as well as Vpx:Vpx interactions. However, no strict correlation was observed between these parameters and the functionality of Vpx, implying that neither properties is absolutely required for this function and indicating that even unstable Vpx mutants may be very efficient in inducing SAMHD1 degradation. Overall, our analysis identifies several Vpx residues required for SAMHD1 degradation and points to a very efficient and plastic mechanism through which Vpx depletes this restriction factor.


Assuntos
HIV-2/metabolismo , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Animais , Antivirais/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Dendríticas/virologia , Células HEK293 , Células HeLa , Humanos , Macrófagos/virologia , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Mutação , Células NIH 3T3 , Ligação Proteica , Proteína 1 com Domínio SAM e Domínio HD , Fatores de Virulência/metabolismo
17.
Cell Signal ; 24(7): 1414-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22406083

RESUMO

Autophagy plays important roles during innate and adaptive immune responses to pathogens, including virus infection. Viruses develop ways to subvert the pathway for their own benefit in order to escape restriction by autophagy, leading to increased viral replication and/or control over apoptosis of their host cells. The effects of HIV infection on the autophagic pathway in host cells have been little documented. Using the susceptible Jurkat cell line and CD4(+) T cells, we studied the relationship of HIV-1 and -2 infections with autophagy. We found that HIV infections significantly increase transcription of ULK1, a member of the autophagy-initiated complex. Two ubiquitin-like conjugation systems, the Atg12 conjugation system and the microtubule-associated protein L chain 3 (LC3) conjugation system that control the elongation of the autophore to form the autophagosome, were activated after HIV infection, with upregulation of Atg12-Atg5 complex and increased transcription of LC3, and formed more autophagosome in infected cells detected using an EM assay. We also found that HIV-1 induced more autophagic death in Jurkat cells relative to HIV-2, and the inhibition of autophagy with 3MA and Beclin-1 knockdown decreased HIV-1 replication significantly. The results indicate that HIV is able to induce the autophagic signaling pathway in HIV-infected host cells, which may be required for HIV infection-mediated apoptotic cell death.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/genética , Infecções por HIV/genética , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Proteínas Reguladoras de Apoptose/genética , Autofagia/efeitos dos fármacos , Proteína 12 Relacionada à Autofagia , Proteína 5 Relacionada à Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Cisteína Endopeptidases/metabolismo , Regulação da Expressão Gênica , HIV-1/metabolismo , HIV-1/patogenicidade , HIV-2/metabolismo , HIV-2/patogenicidade , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Jurkat , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Replicação Viral/efeitos dos fármacos
18.
Virology ; 427(1): 67-75, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22386056

RESUMO

HIV-2 Vpx, a virus-associated accessory protein, is critical for infection of non-dividing myeloid cells. To understand the function of Vpx ubiquitination, interaction with an E3 ubiquitin ligase complex, and ability to overcome an inhibition of reverse transcription, we analyzed Vpx lysine mutants for their function and replication capability in macrophages. Both Wt Vpx and Vpx TA (lysine-less Vpx) localized to the cytoplasm and nucleus in HeLa cells. All HIV-2 Vpx lysine mutants were functional in virion packaging. However, ubiquitination was absent with Vpx TA and Vpx K84A mutants, indicating a lack of ubiquitin on positions K68 and K77. Mutants Vpx K68A and K77A were unable to infect macrophages due to impaired reverse transcription from loss of interaction with the ubiquitin substrate receptor, DCAF1. Even though Vpx K84A lacked ubiquitination, it bound DCAF1, and infected macrophages comparable to Wt Vpx.


Assuntos
Proteínas de Transporte/metabolismo , Infecções por HIV/virologia , HIV-2/metabolismo , Macrófagos/virologia , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitinação , Proteínas Virais Reguladoras e Acessórias/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Células HEK293 , HIV-2/genética , Células HeLa , Humanos , Lisina/genética , Lisina/metabolismo , Macrófagos/metabolismo , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases , Ubiquitina-Proteína Ligases , Proteínas Virais Reguladoras e Acessórias/genética , Vírion/genética , Vírion/metabolismo , Replicação Viral
19.
J Med Primatol ; 41(1): 52-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22017399

RESUMO

BACKGROUND: Although the interactions of cellular cytidine deaminase A3G and viral infection factor (vif) of human immunodeficiency virus (HIV) were reported, regulation of A3G after in vivo HIV infection and disease progression is not known. METHODS: Time courses of plasma virus, CD4(+) T lymphocyte Macaca levels, and concentrations of A3G and vif transcripts were determined in infant macaques infected with HIV-2(287) . These in vivo results were compared with those collected in vitro in HIV-2-infected T cells. RESULTS: Human immunodeficiency virus-infected macaques exhibited plasma viremia (≥10(8) copies/ml) followed by a precipitous CD4(+) T-cell (from 40-70 to ≤5%) decline. An initial increase in A3G transcripts coincides with early increases in virus and vif RNA. As virus load continues to increase, A3G RNA decreases but recovers at a later phase as virus level stabilizes. Pearson correlation analysis revealed strong interactions of A3G-CD4, vif-CD4, and A3G-vif. CONCLUSIONS: There is a time-dependent A3G and vif RNA interaction throughout the course of HIV infection.


Assuntos
Adenosina Desaminase/metabolismo , Produtos do Gene vif/metabolismo , Infecções por HIV/veterinária , HIV-2/metabolismo , Macaca nemestrina , Doenças dos Macacos/virologia , Linfócitos T/virologia , Adenosina Desaminase/genética , Animais , Linhagem Celular , Progressão da Doença , Produtos do Gene vif/genética , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-2/genética , Humanos , Immunoblotting , Doenças dos Macacos/enzimologia , Doenças dos Macacos/genética , Doenças dos Macacos/imunologia , RNA Viral/química , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Linfócitos T/imunologia , Fatores de Tempo , Viremia/veterinária
20.
PLoS One ; 5(3): e9786, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20352106

RESUMO

Human antibody 4E10 targets the highly conserved membrane-proximal external region (MPER) of the HIV-1 transmembrane glycoprotein, gp41, and has extraordinarily broad neutralizing activity. It is considered by many to be a prototype for vaccine development. In this study, we describe four subjects infected with viruses carrying rare MPER polymorphisms associated with resistance to 4E10 neutralization. In one case resistant virus carrying a W680G substitution was transmitted from mother to infant. We used site-directed mutagenesis to demonstrate that the W680G substitution is necessary for conferring the 4E10-resistant phenotype, but that it is not sufficient to transfer the phenotype to a 4E10-sensitive Env. Our third subject carried Envs with a W680R substitution causing variable resistance to 4E10, indicating that residues outside the MPER are required to confer the phenotype. A fourth subject possessed a F673L substitution previously associated with 4E10 resistance. For all three subjects with W680 polymorphisms, we observed additional residues in the MPER that co-varied with position 680 and preserved charged distributions across this region. Our data provide important caveats for vaccine development targeting the MPER. Naturally occurring Env variants described in our study also represent unique tools for probing the structure-function of HIV-1 envelope.


Assuntos
Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/genética , Polimorfismo Genético , Feminino , Produtos do Gene env/metabolismo , Genótipo , HIV-1/metabolismo , HIV-2/metabolismo , Humanos , Lactente , Recém-Nascido , Concentração Inibidora 50 , Funções Verossimilhança , Mães , Mutagênese Sítio-Dirigida , Testes de Neutralização , Fenótipo , Proteínas Recombinantes de Fusão/química , Zâmbia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA