Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Rep ; 36(12): 109735, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34551301

RESUMEN

Subtype C is the most prevalent clade of human immunodeficiency virus type 1 (HIV-1) worldwide. The reasons for this are poorly understood. Here, we demonstrate that a characteristic additional third nuclear factor κB (NF-κB) binding site in the long terminal repeat (LTR) promoter allows subtype C HIV-1 strains to evade restriction by nuclear PYHIN proteins, which sequester the transcription factor Sp1. Further, other LTR alterations are responsible for rare PYHIN resistance of subtype B viruses. Resistance-conferring mutations generally reduce the dependency of HIV-1 on Sp1 for virus production and render LTR transcription highly responsive to stimulation by NF-κB/p65. A third NF-κB binding site increases infectious virus yield in primary CD4+ T cells in an γ-interferon-inducible protein 16 (IFI16)-dependent manner. Comprehensive sequence analyses suggest that the frequency of circulating PYHIN-resistant HIV-1 strains is increasing. Our finding that an additional NF-κB binding site in the LTR confers resistance to nuclear PYHIN proteins helps to explain the dominance of clade C HIV-1 strains.


Asunto(s)
VIH-1/genética , FN-kappa B/química , Proteínas Nucleares/metabolismo , Sitios de Unión , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Susceptibilidad a Enfermedades , Genotipo , Células HEK293 , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Humanos , FN-kappa B/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Secuencias Repetidas Terminales/genética , Replicación Viral
2.
Nat Commun ; 12(1): 4584, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321474

RESUMEN

Interferon-induced transmembrane proteins (IFITMs 1, 2 and 3) can restrict viral pathogens, but pro- and anti-viral activities have been reported for coronaviruses. Here, we show that artificial overexpression of IFITMs blocks SARS-CoV-2 infection. However, endogenous IFITM expression supports efficient infection of SARS-CoV-2 in human lung cells. Our results indicate that the SARS-CoV-2 Spike protein interacts with IFITMs and hijacks them for efficient viral infection. IFITM proteins were expressed and further induced by interferons in human lung, gut, heart and brain cells. IFITM-derived peptides and targeting antibodies inhibit SARS-CoV-2 entry and replication in human lung cells, cardiomyocytes and gut organoids. Our results show that IFITM proteins are cofactors for efficient SARS-CoV-2 infection of human cell types representing in vivo targets for viral transmission, dissemination and pathogenesis and are potential targets for therapeutic approaches.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , Antígenos de Diferenciación/genética , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/metabolismo , Anticuerpos Neutralizantes/farmacología , Antígenos de Diferenciación/metabolismo , Sitios de Unión , COVID-19/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Interferón beta/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/metabolismo , Acoplamiento Viral/efectos de los fármacos
3.
J Virol ; 95(17): e0080821, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34132575

RESUMEN

Simian immunodeficiency virus infecting sooty mangabeys (SIVsmm) has been transmitted to humans on at least nine occasions, giving rise to human immunodeficiency virus type 2 (HIV-2) groups A to I. SIVsmm isolates replicate in human T cells and seem capable of overcoming major human restriction factors without adaptation. However, only groups A and B are responsible for the HIV-2 epidemic in sub-Saharan Africa, and it is largely unclear whether adaptive changes were associated with spread in humans. To address this, we examined the sensitivity of infectious molecular clones (IMCs) of five HIV-2 strains and representatives of five different SIVsmm lineages to various APOBEC3 proteins. We confirmed that SIVsmm strains replicate in human T cells, albeit with more variable replication fitness and frequently lower efficiency than HIV-2 IMCs. Efficient viral propagation was generally dependent on intact vif genes, highlighting the need for counteraction of APOBEC3 proteins. On average, SIVsmm was more susceptible to inhibition by human APOBEC3D, -F, -G, and -H than HIV-2. For example, human APOBEC3F reduced infectious virus yield of SIVsmm by ∼80% but achieved only ∼40% reduction in the case of HIV-2. Functional and mutational analyses of human- and monkey-derived alleles revealed that an R128T polymorphism in APOBEC3F contributes to species-specific counteraction by HIV-2 and SIVsmm Vifs. In addition, a T84S substitution in SIVsmm Vif increased its ability to counteract human APOBEC3F. Altogether, our results confirm that SIVsmm Vif proteins show intrinsic activity against human APOBEC3 proteins but also demonstrate that epidemic HIV-2 strains evolved an increased ability to counteract this class of restriction factors during human adaptation. IMPORTANCE Viral zoonoses pose a significant threat to human health, and it is important to understand determining factors. SIVs infecting great apes gave rise to HIV-1. In contrast, SIVs infecting African monkey species have not been detected in humans, with one notable exception. SIVsmm from sooty mangabeys has crossed the species barrier to humans on at least nine independent occasions and seems capable of overcoming many innate defense mechanisms without adaptation. Here, we confirmed that SIVsmm Vif proteins show significant activity against human APOBEC3 proteins. Our analyses also revealed, however, that different lineages of SIVsmm are significantly more susceptible to inhibition by various human APOBEC3 proteins than HIV-2 strains. Mutational analyses suggest that an R128T substitution in APOBEC3F and a T84S change in Vif contribute to species-specific counteraction by HIV-2 and SIVsmm. Altogether, our results support that epidemic HIV-2 strains acquired increased activity against human APOBEC3 proteins to clear this restrictive barrier.


Asunto(s)
Citosina Desaminasa/metabolismo , Productos del Gen vif/metabolismo , Infecciones por VIH/prevención & control , VIH-2/genética , Interacciones Huésped-Patógeno , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Cercocebus atys , Citosina Desaminasa/genética , Transmisión de Enfermedad Infecciosa/prevención & control , Productos del Gen vif/genética , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Mutación , Síndrome de Inmunodeficiencia Adquirida del Simio/epidemiología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/clasificación , Replicación Viral
4.
Nat Commun ; 12(1): 1282, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627642

RESUMEN

Natural killer (NK) cells play a critical understudied role during HIV infection in tissues. In a natural host of SIV, the African green monkey (AGM), NK cells mediate a strong control of SIVagm infection in secondary lymphoid tissues. We demonstrate that SIVagm infection induces the expansion of terminally differentiated NKG2alow NK cells in secondary lymphoid organs displaying an adaptive transcriptional profile and increased MHC-E-restricted cytotoxicity in response to SIV Env peptides while expressing little IFN-γ. Such NK cell differentiation was lacking in SIVmac-infected macaques. Adaptive NK cells displayed no increased NKG2C expression. This study reveals a previously unknown profile of NK cell adaptation to a viral infection, thus accelerating strategies toward NK-cell directed therapies and viral control in tissues.


Asunto(s)
Células Asesinas Naturales/metabolismo , Ganglios Linfáticos/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Algoritmos , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Chlorocebus aethiops , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Células K562 , Células Asesinas Naturales/citología , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Macaca , Masculino , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Transcriptoma/genética
5.
J Virol ; 95(9)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33597208

RESUMEN

HIV-1 has to overcome physical barriers posed by host cell restriction factors (RFs) for efficient replication. Some RFs, including Trim5α and tetherin, trigger antiviral signaling in addition to directly impairing HIV replication. SERINC5 (S5) is an RF that is incorporated into HIV-1 particles to potently impair their infectivity and is efficiently antagonized by the viral pathogenesis factor Nef. Since effects of S5 on HIV-1 infectivity were mostly studied in reporter cell lines, we analyzed the effects of S5 during infection of primary HIV-1 target cells. In activated CD4+ T lymphocytes, virion incorporation of S5 only moderately impaired virion infectivity and was not associated with altered innate immune recognition. In contrast, in monocyte-derived macrophages, S5 virion incorporation potentiated the production of proinflammatory cytokines with very potent but donor-dependent effects on virion infectivity. Nef counteracted effects of S5 on both cytokine production and virion infectivity. Similar S5-induced cytokine production was observed in immature monocyte-derived dendritic cells. Notably, S5-mediated enhancement of cytokine production was not linked to the efficacy of productive infection and could be overcome by using vesicular stomatitis virus glycoprotein (VSV-G) but not infectivity restriction-insensitive HIV-1 Env for cell entry. Moreover, inhibiting entry of S5-negative HIV-1 ΔNef particles increased proinflammatory cytokine production comparably to virion incorporation of S5. Together, these results describe the sensitization of noninfectious HIV-1 particles to proinflammatory cytokine production by myeloid target cells as an additional and Nef-sensitive activity of S5. Moreover, the study reveals important cell-type and donor-dependent differences in the sensitivity of HIV target cells for antiviral effects of S5.IMPORTANCE SERINC5 (S5) is a host cell restriction factor (RF) that impairs the infectivity of HIV-1 particles in target cell lines. To assess the potential physiological relevance of this restriction, we assessed the effects of S5 on HIV-1 infection of relevant primary human target cells. We found that effects of S5 on infection of CD4+ T lymphocytes were negligible. In myeloid target cells, however, virion incorporation of S5 potently suppressed infectivity and promoted innate immune recognition of HIV-1 particles characterized by proinflammatory cytokine production. Both effects were not observed in cells of all donors analyzed, were exerted independently of one another, and were counteracted by the HIV-1 pathogenesis factor Nef. These results identify the sensitization of HIV-1 particles for innate immune recognition by myeloid target cells as a novel activity of S5 and emphasize the need to study RF function in the context of primary target cells and taking donor variabilities into account.


Asunto(s)
Citocinas/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Interacciones Microbiota-Huesped , Proteínas de la Membrana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células HEK293 , Humanos , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Células Mieloides/inmunología , Virión/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33431697

RESUMEN

GPR15 is a G protein-coupled receptor (GPCR) proposed to play a role in mucosal immunity that also serves as a major entry cofactor for HIV-2 and simian immunodeficiency virus (SIV). To discover novel endogenous GPR15 ligands, we screened a hemofiltrate (HF)-derived peptide library for inhibitors of GPR15-mediated SIV infection. Our approach identified a C-terminal fragment of cystatin C (CysC95-146) that specifically inhibits GPR15-dependent HIV-1, HIV-2, and SIV infection. In contrast, GPR15L, the chemokine ligand of GPR15, failed to inhibit virus infection. We found that cystatin C fragments preventing GPR15-mediated viral entry do not interfere with GPR15L signaling and are generated by proteases activated at sites of inflammation. The antiretroviral activity of CysC95-146 was confirmed in primary CD4+ T cells and is conserved in simian hosts of SIV infection. Thus, we identified a potent endogenous inhibitor of GPR15-mediated HIV and SIV infection that does not interfere with the physiological function of this GPCR.


Asunto(s)
Cistatina C/genética , Infecciones por VIH/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Animales , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Receptores Virales/genética , Transducción de Señal/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Linfocitos T/metabolismo , Linfocitos T/virología , Internalización del Virus
7.
J Am Chem Soc ; 142(40): 17024-17038, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32926779

RESUMEN

Broad-spectrum antivirals are powerful weapons against dangerous viruses where no specific therapy exists, as in the case of the ongoing SARS-CoV-2 pandemic. We discovered that a lysine- and arginine-specific supramolecular ligand (CLR01) destroys enveloped viruses, including HIV, Ebola, and Zika virus, and remodels amyloid fibrils in semen that promote viral infection. Yet, it is unknown how CLR01 exerts these two distinct therapeutic activities. Here, we delineate a novel mechanism of antiviral activity by studying the activity of tweezer variants: the "phosphate tweezer" CLR01, a "carboxylate tweezer" CLR05, and a "phosphate clip" PC. Lysine complexation inside the tweezer cavity is needed to antagonize amyloidogenesis and is only achieved by CLR01. Importantly, CLR01 and CLR05 but not PC form closed inclusion complexes with lipid head groups of viral membranes, thereby altering lipid orientation and increasing surface tension. This process disrupts viral envelopes and diminishes infectivity but leaves cellular membranes intact. Consequently, CLR01 and CLR05 display broad antiviral activity against all enveloped viruses tested, including herpesviruses, Measles virus, influenza, and SARS-CoV-2. Based on our mechanistic insights, we potentiated the antiviral, membrane-disrupting activity of CLR01 by introducing aliphatic ester arms into each phosphate group to act as lipid anchors that promote membrane targeting. The most potent ester modifications harbored unbranched C4 units, which engendered tweezers that were approximately one order of magnitude more effective than CLR01 and nontoxic. Thus, we establish the mechanistic basis of viral envelope disruption by specific tweezers and establish a new class of potential broad-spectrum antivirals with enhanced activity.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Hidrocarburos Aromáticos con Puentes/farmacología , Organofosfatos/farmacología , Proteínas del Envoltorio Viral/efectos de los fármacos , Fosfatasa Ácida/química , Fosfatasa Ácida/metabolismo , Amiloide/antagonistas & inhibidores , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Arginina/química , Betacoronavirus/efectos de los fármacos , Hidrocarburos Aromáticos con Puentes/química , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/virología , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Humanos , Lípidos/química , Lisina/química , Espectroscopía de Resonancia Magnética , Organofosfatos/química , SARS-CoV-2 , Proteínas de Secreción de la Vesícula Seminal/química , Proteínas de Secreción de la Vesícula Seminal/metabolismo , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/metabolismo , Virus Zika/efectos de los fármacos
8.
PLoS Pathog ; 16(8): e1008752, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32760121

RESUMEN

Members of the family of pyrin and HIN domain containing (PYHIN) proteins play an emerging role in innate immunity. While absent in melanoma 2 (AIM2) acts a cytosolic sensor of non-self DNA and plays a key role in inflammasome assembly, the γ-interferon-inducible protein 16 (IFI16) restricts retroviral gene expression by sequestering the transcription factor Sp1. Here, we show that the remaining two human PYHIN proteins, i.e. myeloid cell nuclear differentiation antigen (MNDA) and pyrin and HIN domain family member 1 (PYHIN1 or IFIX) share this antiretroviral function of IFI16. On average, knock-down of each of these three nuclear PYHIN proteins increased infectious HIV-1 yield from human macrophages by more than an order of magnitude. Similarly, knock-down of IFI16 strongly increased virus transcription and production in primary CD4+ T cells. The N-terminal pyrin domain (PYD) plus linker region containing a nuclear localization signal (NLS) were generally required and sufficient for Sp1 sequestration and anti-HIV-1 activity of IFI16, MNDA and PYHIN1. Replacement of the linker region of AIM2 by the NLS-containing linker of IFI16 resulted in a predominantly nuclear localization and conferred direct antiviral activity to AIM2 while attenuating its ability to form inflammasomes. The reverse change caused nuclear-to-cytoplasmic relocalization of IFI16 and impaired its antiretroviral activity but did not result in inflammasome assembly. We further show that the Zn-finger domain of Sp1 is critical for the interaction with IFI16 supporting that pyrin domains compete with DNA for Sp1 binding. Finally, we found that human PYHIN proteins also inhibit Hepatitis B virus and simian vacuolating virus 40 as well as the LINE-1 retrotransposon. Altogether, our data show that IFI16, PYHIN1 and MNDA restrict HIV-1 and other viral pathogens by interfering with Sp1-dependent gene expression and support an important role of nuclear PYHIN proteins in innate antiviral immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Núcleo Celular/metabolismo , Infecciones por VIH/prevención & control , VIH-1/inmunología , Macrófagos/inmunología , Proteínas Nucleares/metabolismo , Factor de Transcripción Sp1/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Núcleo Celular/genética , ADN Viral/genética , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/aislamiento & purificación , Células Hep G2 , Humanos , Inmunidad Innata/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Proteínas Nucleares/genética , Factor de Transcripción Sp1/genética , Replicación Viral
9.
Nat Microbiol ; 5(10): 1247-1261, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32690953

RESUMEN

To avoid innate sensing and immune control, human immunodeficiency virus type 1 (HIV-1) has to prevent the accumulation of viral complementary DNA species. Here, we show that the late HIV-1 accessory protein Vpu hijacks DNA repair mechanisms to promote degradation of nuclear viral cDNA in cells that are already productively infected. Vpu achieves this by interacting with RanBP2-RanGAP1*SUMO1-Ubc9 SUMO E3-ligase complexes at the nuclear pore to reprogramme promyelocytic leukaemia protein nuclear bodies and reduce SUMOylation of Bloom syndrome protein, unleashing end degradation of viral cDNA. Concomitantly, Vpu inhibits RAD52-mediated homologous repair of viral cDNA, preventing the generation of dead-end circular forms of single copies of the long terminal repeat and permitting sustained nucleolytic attack. Our results identify Vpu as a key modulator of the DNA repair machinery. We show that Bloom syndrome protein eliminates nuclear HIV-1 cDNA and thereby suppresses immune sensing and proviral hyper-integration. Therapeutic targeting of DNA repair may facilitate the induction of antiviral immunity and suppress proviral integration replenishing latent HIV reservoirs.


Asunto(s)
Reparación del ADN , Infecciones por VIH/virología , VIH-1/fisiología , Interacciones Huésped-Patógeno , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Inmunidad Innata , Proteínas Reguladoras y Accesorias Virales/metabolismo , Integración Viral , Regulación Viral de la Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Modelos Biológicos , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Reparación del ADN por Recombinación , Sumoilación
10.
Science ; 369(6508): 1249-1255, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32680882

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the current coronavirus disease 2019 (COVID-19) pandemic. A major virulence factor of SARS-CoVs is the nonstructural protein 1 (Nsp1), which suppresses host gene expression by ribosome association. Here, we show that Nsp1 from SARS-CoV-2 binds to the 40S ribosomal subunit, resulting in shutdown of messenger RNA (mRNA) translation both in vitro and in cells. Structural analysis by cryo-electron microscopy of in vitro-reconstituted Nsp1-40S and various native Nsp1-40S and -80S complexes revealed that the Nsp1 C terminus binds to and obstructs the mRNA entry tunnel. Thereby, Nsp1 effectively blocks retinoic acid-inducible gene I-dependent innate immune responses that would otherwise facilitate clearance of the infection. Thus, the structural characterization of the inhibitory mechanism of Nsp1 may aid structure-based drug design against SARS-CoV-2.


Asunto(s)
Betacoronavirus/química , Evasión Inmune , Inmunidad Innata , Biosíntesis de Proteínas , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Betacoronavirus/inmunología , Betacoronavirus/metabolismo , Betacoronavirus/fisiología , Sitios de Unión , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Microscopía por Crioelectrón , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Modelos Moleculares , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/virología , Unión Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , ARN Mensajero/metabolismo , Receptores Inmunológicos , Subunidades Ribosómicas Pequeñas de Eucariotas/química , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , SARS-CoV-2
11.
mBio ; 11(4)2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32665270

RESUMEN

HLA-C-mediated antigen presentation induces the killing of human immunodeficiency virus (HIV)-infected CD4+ T cells by cytotoxic T lymphocytes (CTLs). To evade killing, many HIV-1 group M strains decrease HLA-C surface levels using their accessory protein Vpu. However, some HIV-1 group M isolates lack this activity, possibly to prevent the activation of natural killer (NK) cells. Analyzing diverse primate lentiviruses, we found that Vpu-mediated HLA-C downregulation is not limited to pandemic group M but is also found in HIV-1 groups O and P as well as several simian immunodeficiency viruses (SIVs). We show that Vpu targets HLA-C primarily at the protein level, independently of its ability to suppress NF-κB-driven gene expression, and that in some viral lineages, HLA-C downregulation may come at the cost of efficient counteraction of the restriction factor tetherin. Remarkably, HIV-2, which does not carry a vpu gene, uses its accessory protein Vif to decrease HLA-C surface expression. This Vif activity requires intact binding sites for the Cullin5/Elongin ubiquitin ligase complex but is separable from its ability to counteract APOBEC3G. Similar to HIV-1 Vpu, the degree of HIV-2 Vif-mediated HLA-C downregulation varies considerably among different virus isolates. In agreement with opposing selection pressures in vivo, we show that the reduction of HLA-C surface levels by HIV-2 Vif is accompanied by increased NK cell-mediated killing. In summary, our results highlight the complex role of HLA-C in lentiviral infections and demonstrate that HIV-1 and HIV-2 have evolved at least two independent mechanisms to decrease HLA-C levels on infected cells.IMPORTANCE Genome-wide association studies suggest that HLA-C expression is a major determinant of viral load set points and CD4+ T cell counts in HIV-infected individuals. On the one hand, efficient HLA-C expression enables the killing of infected cells by cytotoxic T lymphocytes (CTLs). On the other hand, HLA-C sends inhibitory signals to natural killer (NK) cells and enhances the infectivity of newly produced HIV particles. HIV-1 group M viruses modulate HLA-C expression using the accessory protein Vpu, possibly to balance CTL- and NK cell-mediated immune responses. Here, we show that the second human immunodeficiency virus, HIV-2, can use its accessory protein Vif to evade HLA-C-mediated restriction. Furthermore, our mutational analyses provide insights into the underlying molecular mechanisms. In summary, our results reveal how the two human AIDS viruses modulate HLA-C, a key component of the antiviral immune response.


Asunto(s)
Evolución Molecular , VIH-1/genética , VIH-2/genética , Antígenos HLA-C/genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Proteínas Reguladoras y Accesorias Virales/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/clasificación , VIH-1/inmunología , VIH-2/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Células Asesinas Naturales/inmunología , Linfocitos T Citotóxicos/inmunología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/inmunología
12.
mBio ; 11(1)2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31937644

RESUMEN

CpG dinucleotide suppression has been reported to allow HIV-1 to evade inhibition by the zinc-finger antiviral protein (ZAP). Here, we show that primate lentiviruses display marked differences in CpG frequencies across their genome, ranging from 0.44% in simian immunodeficiency virus SIVwrc from Western red colobus to 2.3% in SIVmon infecting mona monkeys. Moreover, functional analyses of a large panel of human and simian immunodeficiency viruses revealed that the magnitude of CpG suppression does not correlate with their susceptibility to ZAP. However, we found that the number of CpG dinucleotides within a region of ∼700 bases at the 5' end of the env gene determines ZAP sensitivity of primary HIV-1 strains but not of HIV-2. Increased numbers of CpGs in this region were associated with reduced env mRNA expression and viral protein production. ZAP sensitivity profiles of chimeric simian-human immunodeficiency viruses (SHIVs) expressing different HIV-1 env genes were highly similar to those of the corresponding HIV-1 strains. The frequency of CpGs in the identified env region correlated with differences in clinical progression rates. Thus, the CpG frequency in a specific part of env, rather than the overall genomic CpG content, governs the susceptibility of HIV-1 to ZAP and might affect viral pathogenicity in vivoIMPORTANCE Evasion of the zinc-finger antiviral protein (ZAP) may drive CpG dinucleotide suppression in HIV-1 and many other viral pathogens but the viral determinants of ZAP sensitivity are poorly defined. Here, we examined CpG suppression and ZAP sensitivity in a large number of primate lentiviruses and demonstrate that their genomic frequency of CpGs varies substantially and does not correlate with ZAP sensitivity. We further show that the number of CpG residues in a defined region at the 5' end of the env gene together with structural features plays a key role in HIV-1 susceptibility to ZAP and correlates with differences in clinical progression rates in HIV-1-infected individuals. Our identification of a specific part of env as a major determinant of HIV-1 susceptibility to ZAP restriction provides a basis for future studies of the underlying inhibitory mechanisms and their potential relevance in the pathogenesis of AIDS.


Asunto(s)
Islas de CpG , VIH-1/genética , Proteínas de Unión al ARN/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Genoma Viral , Células HEK293 , VIH-1/patogenicidad , VIH-2/genética , Humanos , Virus de la Inmunodeficiencia de los Simios/genética , Replicación Viral
13.
J Virol ; 93(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31375574

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) has evolved elaborate ways to evade immune cell recognition, including downregulation of classical HLA class I (HLA-I) from the surfaces of infected cells. Recent evidence identified HLA-E, a nonclassical HLA-I, as an important part of the antiviral immune response to HIV-1. Changes in HLA-E surface levels and peptide presentation can prompt both CD8+ T-cell and natural killer (NK) cell responses to viral infections. Previous studies reported unchanged or increased HLA-E levels on HIV-1-infected cells. Here, we examined HLA-E surface levels following infection of CD4+ T cells with primary HIV-1 strains and observed that a subset downregulated HLA-E. Two primary strains of HIV-1 that induced the strongest reduction in surface HLA-E expression were chosen for further testing. Expression of single Nef or Vpu proteins in a T-cell line, as well as tail swap experiments exchanging the cytoplasmic tail of HLA-A2 with that of HLA-E, demonstrated that Nef modulated HLA-E surface levels and targeted the cytoplasmic tail of HLA-E. Furthermore, infection of primary CD4+ T cells with HIV-1 mutants showed that a lack of functional Nef (and Vpu to some extent) impaired HLA-E downmodulation. Taken together, the results of this study demonstrate for the first time that HIV-1 can downregulate HLA-E surface levels on infected primary CD4+ T cells, potentially rendering them less vulnerable to CD8+ T-cell recognition but at increased risk of NKG2A+ NK cell killing.IMPORTANCE For almost two decades, it was thought that HIV-1 selectively downregulated the highly expressed HLA-I molecules HLA-A and HLA-B from the cell surface in order to evade cytotoxic-T-cell recognition, while leaving HLA-C and HLA-E molecules unaltered. It was stipulated that HIV-1 infection thereby maintained inhibition of NK cells via inhibitory receptors that bind HLA-C and HLA-E. This concept was recently revised when a study showed that primary HIV-1 strains reduce HLA-C surface levels, whereas the cell line-adapted HIV-1 strain NL4-3 lacks this ability. Here, we demonstrate that infection with distinct primary HIV-1 strains results in significant downregulation of surface HLA-E levels. Given the increasing evidence for HLA-E as an important modulator of CD8+ T-cell and NKG2A+ NK cell functions, this finding has substantial implications for future immunomodulatory approaches aimed at harnessing cytotoxic cellular immunity against HIV.


Asunto(s)
Regulación de la Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/fisiología , Antígenos de Histocompatibilidad Clase I/genética , Interacciones Huésped-Patógeno/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Biomarcadores , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Membrana Celular/metabolismo , Infecciones por VIH/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunofenotipificación , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Antígenos HLA-E
14.
Cell Host Microbe ; 26(3): 359-368.e8, 2019 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-31447307

RESUMEN

Tetherin is a host defense factor that physically prevents virion release from the plasma membrane. The Nef accessory protein of simian immunodeficiency virus (SIV) engages the clathrin adaptor AP-2 to downregulate tetherin via its DIWK motif. As human tetherin lacks DIWK, antagonism of tetherin by Nef is a barrier to simian-human transmission of non-human primate lentiviruses. To determine the molecular basis for tetherin counteraction, we reconstituted the AP-2 complex with a simian tetherin and SIV Nef and determined its structure by cryoelectron microscopy (cryo-EM). Nef refolds the first α-helix of the ß2 subunit of AP-2 to a ß hairpin, creating a binding site for the DIWK sequence. The tetherin binding site in Nef is distinct from those of most other Nef substrates, including MHC class I, CD3, and CD4 but overlaps with the site for the restriction factor SERINC5. This structure explains the dependence of SIVs on tetherin DIWK and consequent barrier to human transmission.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Antígeno 2 del Estroma de la Médula Ósea/química , Antígeno 2 del Estroma de la Médula Ósea/farmacología , Infecciones por Lentivirus/prevención & control , Infecciones por Lentivirus/transmisión , Zoonosis/virología , Complejo 2 de Proteína Adaptadora/química , Complejo 2 de Proteína Adaptadora/metabolismo , Subunidades beta de Complejo de Proteína Adaptadora/química , Animales , Sitios de Unión , Complejo CD3/metabolismo , Antígenos CD4/metabolismo , Membrana Celular/efectos de los fármacos , Microscopía por Crioelectrón , Regulación hacia Abajo , Productos del Gen nef/química , Productos del Gen nef/metabolismo , Células HEK293 , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Infecciones por Lentivirus/virología , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Cultivo Primario de Células , Conformación Proteica , Conformación Proteica en Hélice alfa , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/metabolismo , Virión/efectos de los fármacos
15.
mBio ; 10(3)2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31213558

RESUMEN

The HIV-1 accessory protein Vpu enhances viral release by counteracting the restriction factor BST-2. Furthermore, Vpu promotes NK cell evasion by downmodulating cell surface NTB-A and PVR, known ligands of the NK cell receptors NTB-A and DNAM-1, respectively. While it has been established that Vpu's transmembrane domain (TMD) is required for the interaction and intracellular sequestration of BST-2, NTB-A, and PVR, it remains unclear how Vpu manages to target these proteins simultaneously. In this study, we show that upon upregulation, BST-2 is preferentially downregulated by Vpu over its other TMD substrates. We found that type I interferon (IFN)-mediated BST-2 upregulation greatly impairs the ability of Vpu to downregulate NTB-A and PVR. Our results suggest that occupation of Vpu by BST-2 affects its ability to downregulate other TMD substrates. Accordingly, knockdown of BST-2 increases Vpu's potency to downmodulate NTB-A and PVR in the presence of type I IFN treatment. Moreover, we show that expression of human BST-2, but not that of the macaque orthologue, decreases Vpu's capacity to downregulate NTB-A. Importantly, we show that type I IFNs efficiently sensitize HIV-1-infected cells to NTB-A- and DNAM-1-mediated direct and antibody-dependent NK cell responses. Altogether, our results reveal that type I IFNs decrease Vpu's polyfunctionality, thus reducing its capacity to protect HIV-1-infected cells from NK cell responses.IMPORTANCE The restriction factor BST-2 and the NK cell ligands NTB-A and PVR are among a growing list of membrane proteins found to be downregulated by HIV-1 Vpu. BST-2 antagonism enhances viral release, while NTB-A and PVR downmodulation contributes to NK cell evasion. However, it remains unclear how Vpu can target multiple cellular factors simultaneously. Here we provide evidence that under physiological conditions, BST-2 is preferentially targeted by Vpu over NTB-A and PVR. Specifically, we show that type I IFNs decrease Vpu's polyfunctionality by upregulating BST-2, thus reducing its capacity to protect HIV-1-infected cells from NK cell responses. This indicates that there is a hierarchy of Vpu substrates upon IFN treatment, revealing that for the virus, targeting BST-2 as part of its resistance to IFN takes precedence over evading NK cell responses. This reveals a potential weakness in HIV-1's immunoevasion mechanisms that may be exploited therapeutically to harness NK cell responses against HIV-1.


Asunto(s)
Antígenos CD/genética , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Interferón Tipo I/farmacología , Células Asesinas Naturales/inmunología , Proteínas Reguladoras y Accesorias Virales/genética , Linfocitos T CD4-Positivos/virología , Regulación hacia Abajo , Proteínas Ligadas a GPI/genética , Células HEK293 , VIH-1 , Proteínas del Virus de la Inmunodeficiencia Humana/inmunología , Humanos , Evasión Inmune , Receptores Virales/genética , Receptores Virales/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Activación Transcripcional , Regulación hacia Arriba , Proteínas Reguladoras y Accesorias Virales/inmunología
16.
Cell Host Microbe ; 25(6): 858-872.e13, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31175045

RESUMEN

The interferon γ-inducible protein 16 (IFI16) is known as immune sensor of retroviral DNA intermediates. We show that IFI16 restricts HIV-1 independently of immune sensing by binding and inhibiting the host transcription factor Sp1 that drives viral gene expression. This antiretroviral activity and ability to bind Sp1 require the N-terminal pyrin domain and nuclear localization of IFI16, but not the HIN domains involved in DNA binding. Highly prevalent clade C HIV-1 strains are more resistant to IFI16 and less dependent on Sp1 than other HIV-1 subtypes. Furthermore, inhibition of Sp1 by IFI16 or pharmacologically by Mithramycin A suppresses reactivation of latent HIV-1 in CD4+ T cells. Finally, IFI16 also inhibits retrotransposition of LINE-1, known to engage Sp1, and murine IFI16 homologs restrict Friend retrovirus replication in mice. Thus, IFI16 restricts retroviruses and retrotransposons by interfering with Sp1-dependent gene expression, and evasion from this restriction may facilitate spread of HIV-1 subtype C.


Asunto(s)
VIH-1/inmunología , Factores Inmunológicos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Transcripción Genética , Activación Viral , Latencia del Virus , Animales , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Genotipo , VIH-1/clasificación , VIH-1/genética , VIH-1/crecimiento & desarrollo , Ratones
17.
PLoS Pathog ; 14(8): e1007269, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30125328

RESUMEN

SERINC5 is a host restriction factor that impairs infectivity of HIV-1 and other primate lentiviruses and is counteracted by the viral accessory protein Nef. However, the importance of SERINC5 antagonism for viral replication and cytopathicity remained unclear. Here, we show that the Nef protein of the highly divergent SIVcol lineage infecting mantled guerezas (Colobus guereza) is a potent antagonist of SERINC5, although it lacks the CD4, CD3 and CD28 down-modulation activities exerted by other primate lentiviral Nefs. In addition, SIVcol Nefs decrease CXCR4 cell surface expression, suppress TCR-induced actin remodeling, and counteract Colobus but not human tetherin. Unlike HIV-1 Nef proteins, SIVcol Nef induces efficient proteasomal degradation of SERINC5 and counteracts orthologs from highly divergent vertebrate species, such as Xenopus frogs and zebrafish. A single Y86F mutation disrupts SERINC5 and tetherin antagonism but not CXCR4 down-modulation by SIVcol Nef, while mutation of a C-proximal di-leucine motif has the opposite effect. Unexpectedly, the Y86F change in SIVcol Nef had little if any effect on viral replication and CD4+ T cell depletion in preactivated human CD4+ T cells and in ex vivo infected lymphoid tissue. However, SIVcol Nef increased virion infectivity up to 10-fold and moderately increased viral replication in resting peripheral blood mononuclear cells (PBMCs) that were first infected with HIV-1 and activated three or six days later. In conclusion, SIVcol Nef lacks several activities that are conserved in other primate lentiviruses and utilizes a distinct proteasome-dependent mechanism to counteract SERINC5. Our finding that evolutionarily distinct SIVcol Nefs show potent anti-SERINC5 activity supports a relevant role of SERINC5 antagonism for viral fitness in vivo. Our results further suggest this Nef function is particularly important for virion infectivity under conditions of limited CD4+ T cell activation.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Productos del Gen nef/fisiología , VIH-1/fisiología , Tejido Linfoide/virología , Proteínas de la Membrana/metabolismo , Replicación Viral/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Colobus/virología , Células HEK293 , Humanos , Células Jurkat , Proteínas de la Membrana/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Virus de la Inmunodeficiencia de los Simios/genética
18.
Cell ; 174(3): 659-671.e14, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30053425

RESUMEN

The HIV accessory protein Nef counteracts immune defenses by subverting coated vesicle pathways. The 3.7 Å cryo-EM structure of a closed trimer of the clathrin adaptor AP-1, the small GTPase Arf1, HIV-1 Nef, and the cytosolic tail of the restriction factor tetherin suggested a mechanism for inactivating tetherin by Golgi retention. The 4.3 Å structure of a mutant Nef-induced dimer of AP-1 showed how the closed trimer is regulated by the dileucine loop of Nef. HDX-MS and mutational analysis were used to show how cargo dynamics leads to alternative Arf1 trimerization, directing Nef targets to be either retained at the trans-Golgi or sorted to lysosomes. Phosphorylation of the NL4-3 M-Nef was shown to regulate AP-1 trimerization, explaining how O-Nefs lacking this phosphosite counteract tetherin but most M-Nefs do not. These observations show how the higher-order organization of a vesicular coat can be allosterically modulated to direct cargoes to distinct fates.


Asunto(s)
Factor de Transcripción AP-1/ultraestructura , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/ultraestructura , Factor 1 de Ribosilacion-ADP/metabolismo , Factor 1 de Ribosilacion-ADP/ultraestructura , Proteínas Adaptadoras del Transporte Vesicular , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/ultraestructura , Clatrina , Aparato de Golgi , Células HEK293 , VIH-1 , Humanos , Transporte de Proteínas/fisiología , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-1/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología
19.
J Virol ; 92(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29743357

RESUMEN

Cellular antiviral proteins interfere with distinct steps of replication cycles of viruses. The galectin 3 binding protein (LGALS3BP, also known as 90K) was previously shown to lower the infectivity of nascent human immunodeficiency virus type 1 (HIV-1) virions when expressed in virus-producing cells. This antiviral effect was accompanied by impaired gp160Env processing and reduced viral incorporation of mature Env glycoproteins. Here, we examined the ability of 90K orthologs from primate species to reduce the particle infectivity of distinct lentiviruses. We show that 90K's ability to diminish the infectivity of lentiviral particles is conserved within primate species, with the notable exception of 90K from rhesus macaque. Comparison of active and inactive 90K orthologs and variants uncovered the fact that inhibition of processing of the HIV-1 Env precursor and reduction of cell surface expression of HIV-1 Env gp120 are required, but not sufficient, for 90K-mediated antiviral activity. Rather, 90K-mediated reduction of virion-associated gp120 coincided with antiviral activity, suggesting that 90K impairs the incorporation of HIV-1 Env into budding virions. We show that a single "humanizing" amino acid exchange in the BTB (broad-complex, tramtrack, and bric-à-brac)/POZ (poxvirus and zinc finger) domain is sufficient to fully rescue the antiviral activity of a shortened version of rhesus macaque 90K, but not that of the full-length protein. Comparison of the X-ray structures of the BTB/POZ domains of 90K from rhesus macaques and humans point toward a slightly larger hydrophobic patch at the surface of the rhesus macaque BTB domain that may modulate a direct interaction with either a second 90K domain or a different protein.IMPORTANCE The cellular 90K protein has been shown to diminish the infectivity of nascent HIV-1 particles. When produced in 90K-expressing cells, particles bear smaller amounts of the HIV-1 Env glycoprotein, which is essential for attaching to and entering new target cells in the subsequent infection round. However, whether the antiviral function of 90K is conserved across primates is unknown. Here, we found that 90K orthologs from most primate species, but, surprisingly, not from rhesus macaques, inhibit HIV-1. The introduction of a single amino acid exchange into a short version of the rhesus macaque 90K protein, consisting of the two intermediate domains of 90K, resulted in full restoration of antiviral activity. Structural elucidation of the respective domain suggests that the absence of antiviral activity in the rhesus macaque factor may be linked to a subtle change in protein-protein interaction.


Asunto(s)
Antígenos de Neoplasias/farmacología , Antivirales/farmacología , Biomarcadores de Tumor/farmacología , Proteínas Portadoras/farmacología , Glicoproteínas/farmacología , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/química , Biomarcadores de Tumor/química , Proteínas Portadoras/química , Productos del Gen env/metabolismo , Glicoproteínas/química , Infecciones por VIH/virología , Humanos , Macaca mulatta , Conformación Proteica , Homología de Secuencia , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Especificidad de la Especie , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
20.
Nat Commun ; 9(1): 1371, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636452

RESUMEN

HIV-1 causes chronic inflammation and AIDS in humans, whereas related simian immunodeficiency viruses (SIVs) replicate efficiently in their natural hosts without causing disease. It is currently unknown to what extent virus-specific properties are responsible for these different clinical outcomes. Here, we incorporate two putative HIV-1 virulence determinants, i.e., a Vpu protein that antagonizes tetherin and blocks NF-κB activation and a Nef protein that fails to suppress T cell activation via downmodulation of CD3, into a non-pathogenic SIVagm strain and test their impact on viral replication and pathogenicity in African green monkeys. Despite sustained high-level viremia over more than 4 years, moderately increased immune activation and transcriptional signatures of inflammation, the HIV-1-like SIVagm does not cause immunodeficiency or any other disease. These data indicate that species-specific host factors rather than intrinsic viral virulence factors determine the pathogenicity of primate lentiviruses.


Asunto(s)
VIH-1/patogenicidad , Especificidad del Huésped , Proteínas del Virus de la Inmunodeficiencia Humana/inmunología , Lentivirus de los Primates/crecimiento & desarrollo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Proteínas Reguladoras y Accesorias Virales/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Secuencia de Aminoácidos , Animales , Antígeno 2 del Estroma de la Médula Ósea/genética , Antígeno 2 del Estroma de la Médula Ósea/inmunología , Complejo CD3/genética , Complejo CD3/inmunología , Chlorocebus aethiops , Femenino , Regulación de la Expresión Génica , VIH-1/crecimiento & desarrollo , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Lentivirus de los Primates/patogenicidad , Activación de Linfocitos , FN-kappa B/genética , FN-kappa B/inmunología , Alineación de Secuencia , Transducción de Señal , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Transcripción Genética , Carga Viral , Proteínas Reguladoras y Accesorias Virales/genética , Virulencia , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA