Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.314
Filtrar
1.
Comput Biol Chem ; 104: 107871, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37084691

RESUMEN

Nef is a small accessory protein pivotal in the HIV-1 viral replication cycle. It is a multifunctional protein and its interactions with kinases in host cells have been well characterized through many in vitro and structural studies. Nef forms a homodimer to activate the kinases and subsequently the phosphorylation pathways. The disruption of its homodimerization represents a valuable approach in the search for novel classes of antiretroviral. However, this research avenue is still underdeveloped as just a few Nef inhibitors have been reported so far, with very limited structural information about their mechanism of action. To address this issue, we have employed an in silico structure-based drug design strategy that combines de novo ligand design with molecular docking and extensive molecular dynamics simulations. Since the Nef pocket involved in homodimerization has high lipophilicity, the initial de novo-designed structures displayed poor drug-likeness and solubility. Taking information from the hydration sites within the homodimerization pocket, structural modifications in the initial lead compound have been introduced to improve the solubility and drug-likeness, without affecting the binding profile. We propose lead compounds that can be the starting point for further optimizations to deliver long-awaited, rationally designed Nef inhibitors.


Asunto(s)
VIH-1 , Simulación del Acoplamiento Molecular , Diseño Asistido por Computadora , Productos del Gen nef , Computadores
2.
Viruses ; 14(12)2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36560778

RESUMEN

Epidemic Kaposi's sarcoma (KS), defined by co-infection with Human Herpes Virus 8 (HHV-8) and the Human Immunodeficiency Virus (HIV), is a major cause of mortality in sub-Saharan Africa. Antiretroviral therapy (ART) significantly reduces the risk of developing KS, and for those with KS, tumors frequently resolve with ART alone. However, for unknown reasons, a significant number of KS cases do not resolve and can progress to death. To explore how HIV responds to ART in the KS tumor microenvironment, we sequenced HIV env-nef found in DNA and RNA isolated from plasma, peripheral blood mononuclear cells, and tumor biopsies, before and after ART, in four Ugandan study participants who had unresponsive or progressive KS after 180-250 days of ART. We performed immunohistochemistry experiments to detect viral proteins in matched formalin-fixed tumor biopsies. Our sequencing results showed that HIV diversity and RNA expression in KS tumors are maintained after ART, despite undetectable plasma viral loads. The presence of spliced HIV transcripts in KS tumors after ART was consistent with a transcriptionally active viral reservoir. Immunohistochemistry staining found colocalization of HIV Nef protein and tissue-resident macrophages in the KS tumors. Overall, our results demonstrated that even after ART reduced plasma HIV viral load to undetectable levels and restored immune function, HIV in KS tumors continues to be transcriptionally and translationally active, which could influence tumor maintenance and progression.


Asunto(s)
Infecciones por VIH , Herpesvirus Humano 8 , Sarcoma de Kaposi , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Humanos , Productos del Gen nef , Herpesvirus Humano 8/genética , VIH/genética , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Leucocitos Mononucleares/patología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , ARN , Microambiente Tumoral
3.
J Virol ; 96(20): e0114822, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36197106

RESUMEN

Long interspersed element type 1 (LINE-1) is the only known type of retroelement that can replicate autonomously, and its retrotransposition activity can trigger interferon (IFN) production. IFN production suppresses the infectivity of exogenous viruses, such as human immunodeficiency virus (HIV). As a counteraction, HIV has been reported to use multiple proteins and mechanisms to suppress LINE-1 replication. However, the mechanisms of HIV-mediated LINE-1 regulation are not fully understood. In this study, we discovered that Nef protein, which is expressed by HIV and is important for HIV pathogenesis, inhibits LINE-1 retrotransposition. Two distinct mechanisms have been uncovered for Nef-induced LINE-1 suppression. Without direct interaction with LINE-1 DNA, Nef potently inhibits the promoter activity of the LINE-1 5'-untranslated region (5'-UTR) and reduces the expression levels of LINE-1 RNA and proteins. Alternatively, although Nef does not bind to the LINE-1 open reading frame 1 protein (ORF1p) or LINE-1 RNA, it significantly compromises the ORF1p-LINE-1 RNA interaction, which is essential for LINE-1 retrotransposition. Both mechanisms can be suppressed by the G2A mutation, which abolishes myristoylation of Nef, suggesting that membrane attachment is essential for Nef to suppress LINE-1. Consequently, through LINE-1 inhibition, Nef downregulates IFN production in host cells. Therefore, our data revealed that Nef is a potent LINE-1 suppressor and an effective innate immune regulator, which not only provides new information on the intricate interaction between HIV, LINE-1, and IFN signaling systems but also strengthens the importance of Nef in HIV infection and highlights the potential of designing novel Nef-targeting anti-HIV drugs. IMPORTANCE Human immunodeficiency viruses are pathogens of AIDS that were first discovered almost 40 years ago and continue to threaten human lives to date. While currently used anti-HIV drugs are sufficient to suppress viral loads in HIV-infected patients, both drug-resistant HIV strains and adverse side effects triggered by the long-term use of these drugs highlight the need to develop novel anti-HIV drugs targeting different viral proteins and/or different steps in viral replication. To achieve this, more information is required regarding HIV pathogenesis and especially its impact on cellular activities in host cells. In this study, we discovered that the Nef protein expressed by HIV potently inhibits LINE-1 retrotransposition. During our attempt to determine the mechanism of Nef-mediated LINE-1 suppression, two additional functions of Nef were uncovered. Nef effectively repressed the promoter activity of LINE-1 5'-UTR and destabilized the interaction between ORF1p and LINE-1 RNA. Consequently, Nef not only compromises LINE-1 replication but also reduces LINE-1-triggered IFN production. The reduction in IFN production, in theory, promotes HIV infectivity. Together with its previously known functions, these findings indicate that Nef is a potential target for the development of novel anti-HIV drugs. Notably, the G2 residue, which has been reported to be essential for most Nef functions, was found to be critical in the regulation of innate immune activation by Nef, suggesting that compromising myristoylation or membrane attachment of Nef may be a good strategy for the inhibition of HIV infection.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , VIH-1 , Humanos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Retroelementos/genética , Productos del Gen nef/genética , Fármacos Anti-VIH/metabolismo , Interferones/metabolismo , ARN/metabolismo , Regiones no Traducidas
4.
Biochem Biophys Res Commun ; 625: 122-127, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35961135

RESUMEN

Human immunodeficiency virus-1 (HIV-1) infection leads to the development of acquired immunodeficiency syndrome (AIDS). To establish a productive infection, HIV-1 hijacks the cellular machinery and modulates various physiological processes to propagate itself. The pathways altered by HIV-1 include cell cycle, autophagy, apoptosis, cell stress pathways, immune response, antiviral response, etc. Zipper interacting protein kinase (ZIPK) is a member of the death-associated protein kinase (DAPK) family of proteins, known to be one of the key regulators of cell death and cell survival pathways. ZIPK is also involved in regulating many cellular processes that are altered during HIV-1 infection; thus, we have explored the functional role of ZIPK in HIV-1 infection. Our results show that ZIPK protein expression is downregulated during HIV-1 infection in Nef dependent manner. Overexpression of ZIPK leads to downregulation in LTR-driven gene expression and virus production, whereas ZIPK knockdown induces viral gene expression and replication. HIV-1 promoter activity is reportedly enhanced by Nef-mediated activation of some transcription factors like NFκB and STAT3. ZIPK is reported to inhibit the STAT3 activity by phosphorylating it at ser-727. Our results show that STAT3 (ser-727) phosphorylation is decreased upon overexpression of Nef with simultaneous downregulation of ZIPK expression. We finally show that HIV-1 Nef interacts with ZIPK and induces its proteasomal degradation. Overall, our data suggests that Nef is involved in downregulation of ZIPK thereby increasing the virus production through rescue of STAT3 activity.


Asunto(s)
Productos del Gen nef , VIH-1 , Proteínas Quinasas Asociadas a Muerte Celular , Productos del Gen nef/fisiología , VIH-1/genética , Humanos , Proteínas Quinasas , Proteínas Virales , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
5.
J Virol ; 96(11): e0017622, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35536019

RESUMEN

Most simian immunodeficiency viruses (SIVs) use Nef to counteract restriction by the tetherin proteins of their nonhuman primate hosts. In addition to counteracting tetherin, SIV Nef has a number of other functions, including the downmodulation of CD3, CD4, and major histocompatibility complex class I (MHC I) molecules from the surface of SIV-infected cells and the enhancement of viral infectivity by preventing the incorporation of SERINC5 into virions. Although these activities require different surfaces of Nef, they can be difficult to separate because of their dependence on similar interactions with AP-1 or AP-2 for clathrin-mediated endocytosis. We previously observed extensive overlap of the SIV Nef residues required for counteracting tetherin and SERINC5. Here, we define substitutions in Nef that separate anti-tetherin activity from SERINC5 antagonism and other activities of Nef. This information was used to engineer an infectious molecular clone of SIV (SIVmac239nefSA) that is sensitive to tetherin but retains CD3, CD4, MHC I, and SERINC5 downmodulation. In primary rhesus macaque CD4+ T cells, SIVmac239nefSA exhibits impaired replication compared to wild-type SIVmac239 under conditions of interferon-induced upregulation of tetherin. These results demonstrate that tetherin antagonism can be separated from other Nef functions and that resistance to tetherin is essential for optimal replication in primary CD4+ T cells. IMPORTANCE Tetherin is an interferon-inducible transmembrane protein that prevents the detachment of enveloped viruses from infected cells by physically tethering nascent virions to cellular membranes. SIV Nef downmodulates simian tetherin to overcome this restriction in nonhuman primate hosts. Nef also enhances virus infectivity by preventing the incorporation of SERINC5 into virions and contributes to immune evasion by downmodulating other proteins from the cell surface. To assess the contribution of tetherin antagonism to virus replication, we engineered an infectious molecular clone of SIV with substitutions in Nef that uncouple tetherin antagonism from other Nef functions. These substitutions impaired virus replication in interferon-treated macaque CD4+ T cells, revealing the impact of tetherin on SIV replication under physiological conditions in primary CD4+ lymphocytes.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea , Productos del Gen nef , Proteínas de la Membrana , Virus de la Inmunodeficiencia de los Simios , Replicación Viral , Animales , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos , Productos del Gen nef/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Interferones/metabolismo , Linfocitos/metabolismo , Linfocitos/virología , Macaca mulatta , Proteínas de la Membrana/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología
6.
PLoS One ; 15(11): e0241667, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33137166

RESUMEN

OBJECTIVE: HIV-associated CNS dysfunction is a significant problem among people with HIV (PWH), who now live longer due to viral suppression from combined anti-retroviral therapy (ART). Over the course of infection, HIV generates toxic viral proteins and induces inflammatory cytokines that have toxic effects on neurons in the CNS. Among these viral proteins, HIV Nef has been found in neurons of postmortem brain specimens from PWH. However, the source of Nef and its impact on neuronal cell homeostasis are still elusive. METHODS AND RESULTS: Here, in using a simian immunodeficiency virus (SIV) infected rhesus macaque model of neuroHIV, we find SIV Nef reactivity in the frontal cortex, hippocampus and cerebellum of SIV-infected animals using immunohistochemistry (IHC). Interestingly, SIV-infected macaques treated with ART also showed frequent Nef positive cells in the cerebellum and hippocampus. Using dual quantitative RNAscope and IHC, we observed cells that were positive for Nef, but were not for SIV RNA, suggesting that Nef protein is present in cells that are not actively infected with SIV. Using cell specific markers, we observed Nef protein in microglia/macrophages and astrocytes. Importantly, we also identified a number of NeuN-positive neurons, which are not permissive to SIV infection, but contained Nef protein. Further characterization of Nef-positive neurons showed caspase 3 activation, indicating late stage apoptosis in the CNS neurons. CONCLUSIONS: Our results suggest that regardless of ART status, Nef is expressed in the brain of SIV infected macaques and may contribute to neurological complications seen in PWH.


Asunto(s)
Cerebelo/metabolismo , Productos del Gen nef/genética , Hipocampo/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Animales , Cerebelo/virología , Productos del Gen nef/metabolismo , Hipocampo/virología , Macaca mulatta , Masculino , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo
7.
PLoS One ; 15(8): e0225420, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32764749

RESUMEN

The H196 residue in SIVmac239 Nef is conserved across the majority of HIV and SIV isolates, lies immediately adjacent to the AP-2 (adaptor protein 2) binding di-leucine domain (ExxxLM195), and is critical for several described AP-2 dependent Nef functions, including the downregulation of tetherin (BST-2/CD317), CD4, and others. Surprisingly, many stocks of the closely related SIVmac251 swarm virus harbor a nef allele encoding a Q196. In SIVmac239, this variant is associated with loss of multiple AP-2 dependent functions. Publicly available sequences for SIVmac251 stocks were mined for variants linked to Q196 that might compensate for functional defects associated with this residue. Variants were engineered into the SIVmac239 backbone and in Nef expression plasmids and flow cytometry was used to examine surface tetherin expression in primary CD4 T cells and surface CD4 expression in SupT1 cells engineered to express rhesus CD4. We found that SIVmac251 stocks that encode a Q196 residue in Nef uniformly also encode an upstream R191 residue. We show that R191 restores the ability of Nef to downregulate tetherin in the presence of Q196 and has a similar but less pronounced impact on CD4 expression. However, a published report showed Q196 commonly evolves to H196 in vivo, suggesting a fitness cost. R191 may represent compensatory evolution to restore the ability to downregulate tetherin lost in viruses harboring Q196.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Proteínas Reguladoras y Accesorias Virales/genética , Animales , Antígenos CD/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/genética , Linfocitos T CD4-Positivos/metabolismo , Proteínas Ligadas a GPI/metabolismo , Productos del Gen nef/metabolismo , Macaca mulatta/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas Reguladoras y Accesorias Virales/metabolismo
8.
Eur J Med Chem ; 204: 112644, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32738412

RESUMEN

Natural products, as an ideal starting point for molecular design, play a pivotal role in drug discovery; however, ambiguous targets and mechanisms have limited their in-depth research and applications in a global dimension. In-silico target prediction methods have become an alternative to target identification experiments due to the high accuracy and speed, but most studies only use a single prediction method, which may reduce the accuracy and reliability of the prediction. Here, we firstly presented a combinatorial target screening strategy to facilitate multi-target screening of natural products considering the characteristics of diverse in-silico target prediction methods, which consists of ligand-based online approaches, consensus SAR modelling and target-specific re-scoring function modelling. To validate the practicability of the strategy, natural product neferine, a bisbenzylisoquinoline alkaloid isolated from the lotus seed, was taken as an example to illustrate the screening process and a series of corresponding experiments were implemented to explore the pharmacological mechanisms of neferine. The proposed computational method could be used for a complementary hypothesis generation and rapid analysis of potential targets of natural products.


Asunto(s)
Productos Biológicos/farmacología , Técnicas Químicas Combinatorias , Sustancias Macromoleculares/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/efectos de los fármacos , Animales , Productos Biológicos/química , Productos del Gen nef/efectos de los fármacos , Humanos , Ligandos , Sustancias Macromoleculares/química , Ratones , Ratones Desnudos , Reproducibilidad de los Resultados , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Ensayos Antitumor por Modelo de Xenoinjerto
9.
PLoS Pathog ; 16(4): e1008487, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32302364

RESUMEN

Most simian immunodeficiency viruses use Nef to counteract the tetherin proteins of their nonhuman primate hosts. Nef also downmodulates cell-surface CD4 and MHC class I (MHC I) molecules and enhances viral infectivity by counteracting SERINC5. We previously demonstrated that tetherin antagonism by SIV Nef is genetically separable from CD4- and MHC I-downmodulation. Here we show that disruption of tetherin antagonism by Nef impairs virus replication during acute SIV infection of rhesus macaques. A combination of mutations was introduced into the SIVmac239 genome resulting in three amino acid substitutions in Nef that impair tetherin antagonism, but not CD3-, CD4- or MHC I-downmodulation. Further characterization of this mutant (SIVmac239AAA) revealed that these changes also result in partial sensitivity to SERINC5. Separate groups of four rhesus macaques were infected with either wild-type SIVmac239 or SIVmac239AAA, and viral RNA loads in plasma and sequence changes in the viral genome were monitored. Viral loads were significantly lower during acute infection in animals infected with SIVmac239AAA than in animals infected with wild-type SIVmac239. Sequence analysis of the virus population in plasma confirmed that the substitutions in Nef were retained during acute infection; however, changes were observed by week 24 post-infection that fully restored anti-tetherin activity and partially restored anti-SERINC5 activity. These observations reveal overlap in the residues of SIV Nef required for counteracting tetherin and SERINC5 and selective pressure to overcome these restriction factors in vivo.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/virología , Productos del Gen nef , Macaca mulatta , Proteínas de la Membrana/metabolismo , ARN Viral/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo , Carga Viral , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores
10.
Elife ; 92020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32119644

RESUMEN

HIV-1 Vpr is necessary for maximal HIV infection and spread in macrophages. Evolutionary conservation of Vpr suggests an important yet poorly understood role for macrophages in HIV pathogenesis. Vpr counteracts a previously unknown macrophage-specific restriction factor that targets and reduces the expression of HIV Env. Here, we report that the macrophage mannose receptor (MR), is a restriction factor targeting Env in primary human monocyte-derived macrophages. Vpr acts synergistically with HIV Nef to target distinct stages of the MR biosynthetic pathway and dramatically reduce MR expression. Silencing MR or deleting mannose residues on Env rescues Env expression in HIV-1-infected macrophages lacking Vpr. However, we also show that disrupting interactions between Env and MR reduces initial infection of macrophages by cell-free virus. Together these results reveal a Vpr-Nef-Env axis that hijacks a host mannose-MR response system to facilitate infection while evading MR's normal role, which is to trap and destroy mannose-expressing pathogens.


Human cells have defense mechanisms against viral infection known as restriction factors. These are proteins that break down parts of a virus including its DNA or proteins. To evade these defenses, viruses in turn make proteins that block or break down restriction factors. This battle between human and viral proteins determines which types of cells are infected and how quickly a virus can multiply and spread to new cells. HIV produces a protein called Vpr that counteracts a restriction factor found in immune cells called macrophages. However, the identity of the restriction factor targeted by Vpr is a mystery. When Vpr is missing, this unknown restriction factor breaks down a virus protein called Env. Env is a glycoprotein, which is a protein with sugars attached. When Env levels are low, HIV cannot spread to other cells and multiply. Identifying the restriction factor that breaks down Env may lead to new ways of treating and preventing HIV infections. Now, Lubow et al. reveal that the unknown restriction factor in macrophages is a protein called the mannose receptor. This protein binds and destroys proteins containing mannose, a type of sugar found on bacteria and some viruses. The experiments revealed that the mannose receptor grabs mannose on the HIV protein Env. This causes Env to be broken down and stops HIV from spreading. Lubow et al. also find that Vpr works with another protein produced by HIV called Nef to reduce the number of mannose receptors on macrophages. The two proteins do this by targeting different steps in the assembly of mannose receptors, allowing the virus to multiply and spread more efficiently. The experiments suggest that drugs that simultaneously block Vpr and Nef might prevent or suppress HIV infections. More studies are needed to develop and test potential HIV-treatments targeting Vpr and Nef.


Asunto(s)
VIH-1/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Lectinas de Unión a Manosa/metabolismo , Receptores de Superficie Celular/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen env/metabolismo , Productos del Gen nef/metabolismo , VIH-1/fisiología , Humanos , Receptor de Manosa , Unión Proteica , Replicación Viral
11.
Cell Rep ; 30(7): 2261-2274.e7, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075764

RESUMEN

The inability of Nef to downmodulate the CD3-T cell receptor (TCR) complex distinguishes HIV-1 from other primate lentiviruses and may contribute to its high virulence. However, the role of this Nef function in virus-mediated immune activation and pathogenicity remains speculative. Here, we selectively disrupted this Nef activity in SIVmac239 and analyzed the consequences for the virological, immunological, and clinical outcome of infection in rhesus macaques. The inability to downmodulate CD3-TCR does not impair viral replication during acute infection but is associated with increased immune activation and antiviral gene expression. Subsequent early reversion in three of six animals suggests strong selective pressure for this Nef function and is associated with high viral loads and progression to simian AIDS. In the absence of reversions, however, viral replication and the clinical course of infection are attenuated. Thus, Nef-mediated downmodulation of CD3 dampens the inflammatory response to simian immunodeficiency virus (SIV) infection and seems critical for efficient viral immune evasion.


Asunto(s)
Evasión Inmune/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Femenino , Productos del Gen nef , Inflamación/inmunología , Inflamación/patología , Macaca mulatta , Masculino , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Virus de la Inmunodeficiencia de los Simios/inmunología
13.
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
14.
J Immunol ; 202(12): 3349-3358, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31043477

RESUMEN

Similar to host proteins, N-myristoylation occurs for viral proteins to dictate their pathological function. However, this lipid-modifying reaction creates a novel class of "lipopeptide" Ags targeted by host CTLs. The primate MHC class I-encoded protein, Mamu-B*098, was previously shown to bind N-myristoylated 5-mer peptides. Nevertheless, T cells exist that recognize even shorter lipopeptides, and much remains to be elucidated concerning the molecular mechanisms of lipopeptide presentation. We, in this study, demonstrate that the MHC class I allele, Mamu-B*05104, binds the N-myristoylated 4-mer peptide (C14-Gly-Gly-Ala-Ile) derived from the viral Nef protein for its presentation to CTLs. A phylogenetic tree analysis indicates that these classical MHC class I alleles are not closely associated; however, the high-resolution x-ray crystallographic analyses indicate that both molecules share lipid-binding structures defined by the exceptionally large, hydrophobic B pocket to accommodate the acylated glycine (G1) as an anchor. The C-terminal isoleucine (I4) of C14-Gly-Gly-Ala-Ile anchors at the F pocket, which is distinct from that of Mamu-B*098 and is virtually identical to that of the peptide-presenting MHC class I molecule, HLA-B51. The two central amino acid residues (G2 and A3) are only exposed externally for recognition by T cells, and the methyl side chain on A3 constitutes a major T cell epitope, underscoring that the epitopic diversity is highly limited for lipopeptides as compared with that for MHC class I-presented long peptides. These structural features suggest that lipopeptide-presenting MHC class I alleles comprise a distinct MHC class I subset that mediates an alternative pathway for CTL activation.


Asunto(s)
Autoantígenos/metabolismo , Epítopos de Linfocito T/metabolismo , Productos del Gen nef/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Lipopéptidos/metabolismo , Péptidos/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Presentación de Antígeno , Autoantígenos/química , Autoantígenos/inmunología , Cristalografía por Rayos X , Epítopos de Linfocito T/inmunología , Productos del Gen nef/química , Productos del Gen nef/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Lipopéptidos/química , Lipopéptidos/inmunología , Activación de Linfocitos , Ácido Mirístico/química , Péptidos/química , Péptidos/inmunología , Filogenia , Primates
15.
AIDS ; 33(6): 953-964, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30946149

RESUMEN

OBJECTIVE: HIV-infected patients receiving antiretroviral treatment (ART) often present adipose tissue accumulation and/or redistribution. adipose tissue has been shown to be an HIV/SIV reservoir and viral proteins as Tat or Nef can be released by infected immune cells and exert a bystander effect on adipocytes or precursors. Our aim was to demonstrate that SIV/HIV infection per se could alter adipose tissue structure and/or function. DESIGN: Morphological and functional alterations of subcutaneous (SCAT) and visceral adipose tissue (VAT) were studied in SIV-infected macaques and HIV-infected ART-controlled patients. To analyze the effect of Tat or Nef, we used human adipose stem cells (ASCs) issued from healthy donors, and analyzed adipogenesis and extracellular matrix component production using two dimensional (2D) and three-dimensional (3D) culture models. METHODS: Adipocyte size and index of fibrosis were determined on Sirius red-stained adipose tissue samples. Proliferating and adipocyte 2D-differentiating or 3D-differentiating ASCs were treated chronically with Tat or Nef. mRNA, protein expression and secretion were examined by RT-PCR, western-blot and ELISA. RESULTS: SCAT and VAT from SIV-infected macaques displayed small adipocytes, decreased adipogenesis and severe fibrosis with collagen deposition. SCAT and VAT from HIV-infected ART-controlled patients presented similar alterations. In vitro, Tat and/or Nef induced a profibrotic phenotype in undifferentiated ASCs and altered adipogenesis and collagen production in adipocyte-differentiating ASCs. CONCLUSION: We demonstrate here a specific role for HIV/SIV infection per se on adipose tissue fibrosis and adipogenesis, probably through the release of viral proteins, which could be involved in adipose tissue dysfunction contributing to cardiometabolic alterations of HIV-infected individuals.


Asunto(s)
Adipogénesis/efectos de los fármacos , Tejido Adiposo/patología , Fibrosis/patología , Infecciones por VIH/patología , VIH/crecimiento & desarrollo , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Adulto , Animales , Células Cultivadas , Femenino , Productos del Gen nef/metabolismo , Productos del Gen tat/metabolismo , Interacciones Huésped-Patógeno , Humanos , Macaca fascicularis , Masculino , Persona de Mediana Edad , Síndrome de Inmunodeficiencia Adquirida del Simio/patología
16.
J Virol ; 93(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30541854

RESUMEN

Approximately 50% of rhesus macaques (RMs) expressing the major histocompatibility complex class I (MHC-I) allele Mamu-B*08 spontaneously control chronic-phase viremia after infection with the pathogenic simian immunodeficiency virus mac239 (SIVmac239) clone. CD8+ T-cell responses in these animals are focused on immunodominant Mamu-B*08-restricted SIV epitopes in Vif and Nef, and prophylactic vaccination with these epitopes increases the incidence of elite control in SIVmac239-infected Mamu-B*08-positive (Mamu-B*08+ ) RMs. Here we evaluated if robust vaccine-elicited CD8+ T-cell responses against Vif and Nef can prevent systemic infection in Mamu-B*08+ RMs following mucosal SIV challenges. Ten Mamu-B*08+ RMs were vaccinated with a heterologous prime/boost/boost regimen encoding Vif and Nef, while six sham-vaccinated MHC-I-matched RMs served as the controls for this experiment. Vaccine-induced CD8+ T cells against Mamu-B*08-restricted SIV epitopes reached high frequencies in blood but were present at lower levels in lymph node and gut biopsy specimens. Following repeated intrarectal challenges with SIVmac239, all control RMs became infected by the sixth SIV exposure. By comparison, four vaccinees were still uninfected after six challenges, and three of them remained aviremic after 3 or 4 additional challenges. The rate of SIV acquisition in the vaccinees was numerically lower (albeit not statistically significantly) than that in the controls. However, peak viremia was significantly reduced in infected vaccinees compared to control animals. We found no T-cell markers that distinguished vaccinees that acquired SIV infection from those that did not. Additional studies will be needed to validate these findings and determine if cellular immunity can be harnessed to prevent the establishment of productive immunodeficiency virus infection.IMPORTANCE It is generally accepted that the antiviral effects of vaccine-induced classical CD8+ T-cell responses against human immunodeficiency virus (HIV) are limited to partial reductions in viremia after the establishment of productive infection. Here we show that rhesus macaques (RMs) vaccinated with Vif and Nef acquired simian immunodeficiency virus (SIV) infection at a lower (albeit not statistically significant) rate than control RMs following repeated intrarectal challenges with a pathogenic SIV clone. All animals in the present experiment expressed the elite control-associated major histocompatibility complex class I (MHC-I) molecule Mamu-B*08 that binds immunodominant epitopes in Vif and Nef. Though preliminary, these results provide tantalizing evidence that the protective efficacy of vaccine-elicited CD8+ T cells may be greater than previously thought. Future studies should examine if vaccine-induced cellular immunity can prevent systemic viral replication in RMs that do not express MHC-I alleles associated with elite control of SIV infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Productos del Gen nef/inmunología , Productos del Gen vif/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Epítopos de Linfocito T/inmunología , Productos del Gen nef/administración & dosificación , Productos del Gen vif/administración & dosificación , Antígenos de Histocompatibilidad Clase I/inmunología , Macaca mulatta , Vacunación , Vacunas Virales/inmunología , Viremia/inmunología
18.
Traffic ; 20(3): 202-212, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30569492

RESUMEN

When studying how HIV-1 Nef can promote packaging of the proinflammatory transmembrane protease TACE (tumor necrosis factor-α converting enzyme) into extracellular vesicles (EVs) we have revealed a novel tyrosine kinase-regulated unconventional protein secretion (UPS) pathway for TACE. When TACE was expressed without its trafficking cofactor iRhom allosteric Hck activation by Nef triggered translocation of TACE into EVs. This process was insensitive to blocking of classical secretion by inhibiting endoplasmic reticulum (ER) to Golgi transport, and involved a distinct form of TACE devoid of normal glycosylation and incompletely processed for prodomain removal. Like most other examples of UPS this process was Golgi reassembly stacking protein (GRASP)-dependent but was not associated with ER stress. These data indicate that Hck-activated UPS provides an alternative pathway for TACE secretion that can bypass iRhom-dependent ER to Golgi transfer, and suggest that tyrosine phosphorylation might have a more general role in regulating UPS.


Asunto(s)
Proteína ADAM17/metabolismo , Vesículas Extracelulares/metabolismo , Vías Secretoras , Productos del Gen nef/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-hck/metabolismo
19.
Mol Biotechnol ; 60(11): 773-782, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30167966

RESUMEN

Eukaryotic cells constitutively produce nanovesicles of 50-150 nm of diameter, referred to as exosomes, upon release of the contents of multivesicular bodies (MVBs). We recently characterized a novel, exosome-based way to induce cytotoxic T lymphocyte (CTL) immunization against full-length antigens. It is based on DNA vectors expressing products of fusion between the exosome-anchoring protein Nef mutant (Nefmut) with the antigen of interest. The strong efficiency of Nefmut to accumulate in MVBs results in the production of exosomes incorporating huge amounts of the desired antigen. When translated in animals, the injection of Nefmut-based DNA vectors generates engineered exosomes whose internalization in antigen-presenting cells induces cross-priming and antigen-specific CTL immunity. Here, we describe the molecular strategies we followed to produce DNA vectors aimed at generating immunogenic exosomes potentially useful to elicit a CTL immune response against antigens expressed by the etiologic agents of major chronic viral infections, i.e., HIV-1, HBV, and the novel tumor-associated antigen HOXB7. Unique methods intended to counteract intrinsic RNA instability and nuclear localization of the antigens have been developed. The success we met with the production of these engineered exosomes opens the way towards pre-clinic experimentations devoted to the optimization of new vaccine candidates against major infectious and tumor pathologies.


Asunto(s)
Exosomas/genética , Vectores Genéticos/administración & dosificación , Linfocitos T Citotóxicos/inmunología , Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Exosomas/inmunología , Productos del Gen nef/genética , Vectores Genéticos/inmunología , Células HEK293 , Hepatitis B/tratamiento farmacológico , Humanos , Neoplasias/tratamiento farmacológico , Vacunas/inmunología
20.
Sensors (Basel) ; 18(8)2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30071687

RESUMEN

A Q-body capable of detecting target molecules in solutions could serve as a simple molecular detection tool. The position of the fluorescent dye in a Q-body affects sensitivity and therefore must be optimized. This report describes the development of Nef Q-bodies that recognize Nef protein, one of the human immunodeficiency virus (HIV)'s gene products, in which fluorescent dye molecules were placed at various positions using an in vivo unnatural amino acid incorporation system. A maximum change in fluorescence intensity of 2-fold was observed after optimization of the dye position. During the process, some tryptophan residues of the antibody were found to quench the fluorescence. Moreover, analysis of the epitope indicated that some amino acid residues of the antigen located near the epitope affected the fluorescence intensity.


Asunto(s)
Aminoácidos/análisis , Aminoácidos/química , Productos del Gen nef/química , Antígenos VIH/química , Secuencia de Aminoácidos , Animales , Epítopos/análisis , Epítopos/química , Fluorescencia , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/química , Productos del Gen nef/análisis , Antígenos VIH/análisis , Humanos , Ratones , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...