Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1225055, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38078012

RESUMEN

Endometriosis is a benign gynecological disease in which eutopic endometrial tissue composed of glands and stroma grow within the pelvic cavity. The disease affects females of reproductive age and is characterized by pelvic pain, infertility and reduced quality of life. The majority of pharmacologic treatment modalities for endometriosis focus on suppression of estradiol production and/or action; an approach associated with adverse side effects. c-MYC is elevated in eutopic endometrium and endometriotic lesion tissue in patients with endometriosis and the disease shares many similar pathological characteristics with that of endometrial carcinoma. While targeting of c-MYC with Omomyc has recently gained substantial interest in the field of cancer research, there has been no recent attempt to evaluate the potential utility in targeting c-MYC for endometriosis treatment. The following perspective article compares the similarities between endometriosis and endometrial cancer and presents preliminary data suggesting that targeting c-MYC with Omomyc reduces endometriotic cell proliferation and viability in vitro. Future application of targeting c-MYC in endometriosis treatment and potential pros and cons are then discussed.

2.
bioRxiv ; 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37547007

RESUMEN

The open reading frame 3a (ORF3a) is an accessory transmembrane protein that is important to the pathogenicity of SARS-CoV-2. The cytoplasmic domain of ORF3a has three canonical tyrosine-based sorting signals (YxxΦ; where x is any amino acid and Φ is a hydrophobic amino acid with a bulky -R group). They have been implicated in the trafficking of membrane proteins to the cell plasma membrane and to intracellular organelles. Previous studies have indicated that mutation of the 160YSNV163 motif abrogated plasma membrane expression and inhibited ORF3a-induced apoptosis. However, two additional canonical tyrosine-based sorting motifs (211YYQL213, 233YNKI236) exist in the cytoplasmic domain of ORF3a that have not been assessed. We removed all three potential tyrosine-based motifs and systematically restored them to assess the importance of each motif or combination of motifs that restored efficient trafficking to the cell surface and lysosomes. Our results indicate that the YxxΦ motif at position 160 was insufficient for the trafficking of ORF3a to the cell surface. Our studies also showed that ORF3a proteins with an intact YxxΦ at position 211 or at 160 and 211 were most important. We found that ORF3a cell surface expression correlated with the co-localization of ORF3a with LAMP-1 near the cell surface. These results suggest that YxxΦ motifs within the cytoplasmic domain may act cooperatively in ORF3a transport to the plasma membrane and endocytosis to lysosomes. Further, our results indicate that certain tyrosine mutants failed to activate caspase 3 and did not correlate with autophagy functions associated with this protein.

3.
Microbiol Spectr ; 11(1): e0370722, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36622177

RESUMEN

The SARS-CoV-2 virion is composed of four structural proteins: spike (S), nucleocapsid (N), membrane (M), and envelope (E). E spans the membrane a single time and is the smallest, yet most enigmatic of the structural proteins. E is conserved among coronaviruses and has an essential role in virus-mediated pathogenesis. We found that ectopic expression of E had deleterious effects on the host cell as it activated stress responses, leading to LC3 lipidation and phosphorylation of the translation initiation factor eIF2α that resulted in host translational shutoff. During infection E is highly expressed, although only a small fraction is incorporated into virions, suggesting that E activity is regulated and harnessed by the virus to its benefit. Consistently, we found that proteins from heterologous viruses, such as the γ1 34.5 protein of herpes simplex virus 1, prevented deleterious effects of E on the host cell and allowed for E protein accumulation. This observation prompted us to investigate whether other SARS-CoV-2 structural proteins regulate E. We found that the N and M proteins enabled E protein accumulation, whereas S did not. While γ1 34.5 protein prevented deleterious effects of E on the host cells, it had a negative effect on SARS-CoV-2 replication. The negative effect of γ1 34.5 was most likely associated with failure of SARS-CoV-2 to divert the translational machinery and with deregulation of autophagy. Overall, our data suggest that SARS-CoV-2 causes stress responses and subjugates these pathways, including host protein synthesis (phosphorylated eIF2α) and autophagy, to support optimal virus replication. IMPORTANCE In late 2019, a new ß-coronavirus, SARS-CoV-2, entered the human population causing a pandemic that has resulted in over 6 million deaths worldwide. Although closely related to SARS-CoV, the mechanisms of SARS-CoV-2 pathogenesis are not fully understood. We found that ectopic expression of the SARS-CoV-2 E protein had detrimental effects on the host cell, causing metabolic alterations, including shutoff of protein synthesis and mobilization of cellular resources through autophagy activation. Coexpression of E with viral proteins known to subvert host antiviral responses such as autophagy and translational inhibition, either from SARS-CoV-2 or from heterologous viruses, increased cell survival and E protein accumulation. However, such factors were found to negatively impact SARS-CoV-2 infection, as autophagy contributes to formation of viral membrane factories and translational control offers an advantage for viral gene expression. Overall, SARS-CoV-2 has evolved mechanisms to harness host functions that are essential for virus replication.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Autofagia , Procesamiento Proteico-Postraduccional , SARS-CoV-2/metabolismo , Proteínas Virales/genética
4.
J Virol ; 95(8)2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33536165

RESUMEN

Previously, we showed that the presence of the herpes simplex virus type 1 (HSV-1) gD glycoprotein but not gB potently restricted HIV-1 particle infectivity. This restriction was characterized by incorporation of HSV-1 gD and the exclusion of the HIV-1 gp120/gp41 from budding virus particles. To determine the structural domains involved in gD restriction of HIV-1, a series of deletion mutants and chimeric proteins between gD and the non-restrictive gB were generated. Our results show that deletion of the cytoplasmic tail domain (CTD) of gD or that replacement of the transmembrane domain (TMD) with the TMD from gB slightly reduced restriction activity. However, replacement of the gD CTD with that of gB resulted in lower cell surface expression, significantly less incorporation into HIV-1 particles, and inefficient restriction of the release of infectious HIV-1. Analysis of gB/gD chimeric proteins revealed that removal of the gB CTD or replacement with gD CTD resulted in enhanced surface expression and an increase in restriction activity. Finally, we show that expression of gD without other HSV-1 proteins resulted in gD fractionation into detergent resistant membranes (DRM) and that gD co-localized with the raft marker GM1, which may partially explain its incorporation into budding virus particles. Taken together, our results suggest that expression of gD at the cell surface is likely a major factor but that other intrinsic properties are also involved in the gD-mediated restriction of HIV-1 particle infectivity.IMPORTANCE Previously, we showed that unlike the HSV-1, the presence of the gD glycoprotein in virus producer cells but not gB potently restricted HIV-1 particle infectivity. To better understand the relationship between cell surface expression, virus incorporation and restriction of HIV-1, we analyzed a series of deletion mutants and chimeric proteins in which domains of gD and gB were swapped. Our results indicate that: a) gD/gB chimeras having the cytoplasmic domain (CTD) of gB significantly reduced cell surface expression, release from cells, incorporation into virus, and reduced HIV-1 restriction; b) removal of the gB CTD or replacement with the gD CTD resulted in better surface expression, incorporation into HIV-1, and enhanced restriction; and c) the transmembrane domain of gB can influence transport and ultimately effect incorporation of gB into HIV-1. Overall, these data support a role for gD surface expression as crucial to restriction of infectious HIV-1 release.

5.
Retrovirology ; 16(1): 9, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940160

RESUMEN

BACKGROUND: We previously showed that the gM of HSV-1 could restrict the release of infectious HIV-1 from cells. In this study, we analyzed if the four HSV-1 glycoproteins (gD, gB, and gH/gL), which are the minimum glycoproteins required for HSV-1 entry, restricted the release of infectious HIV-1. RESULTS: Of these four glycoproteins, gD and gH/gL restricted the production of infectious HIV-1 from cells transfected with an infectious molecular clone of HIV-1 (strain NL4-3) while gB had no significant effect. Pulse-chase analyses indicated that gD did not affect the biosynthesis and processing of gp160 into gp120/gp41, the transport of the gp120/gp41 to the cell surface, or the release of HIV-1 particles from the cell surface. Our analyses revealed that gD was incorporated into HIV-1 virus particles while gp120/gp41 was excluded from released virus particles. Truncated mutants of gD revealed that the cytoplasmic domain was dispensable but that a membrane bound gD was required for the restriction of release of infectious HIV-1. Finally, cell lines expressing gD also potently restricted the release of infectious virus. CONCLUSIONS: Due to its ability to exclude HIV-1 gp120/gp41 from maturing virus, gD may provide a useful tool in deciphering mechanisms of Env incorporation into maturing virus particles.


Asunto(s)
VIH-1/fisiología , Herpesvirus Humano 1/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Línea Celular , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/metabolismo , Proteínas gp160 de Envoltorio del VIH/genética , Proteínas gp160 de Envoltorio del VIH/metabolismo , Herpesvirus Humano 1/genética , Humanos , Glicoproteínas de Membrana/genética , Proteínas del Envoltorio Viral/genética
6.
Brain Behav Immun ; 71: 37-51, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29729322

RESUMEN

Methamphetamine (METH) abuse is common among individuals infected with HIV-1 and has been shown to affect HIV replication and pathogenesis. These HIV-1 infected individuals also exhibit greater neuronal injury and higher cognitive decline. HIV-1 proteins, specifically gp120 and HIV-1 Tat, have been earlier shown to affect neurocognition. HIV-1 Tat, a viral protein released early during HIV-1 replication, contributes to HIV-associated neurotoxicity through various mechanisms including production of pro-inflammatory cytokines, reactive oxygen species and dysregulation of neuroplasticity. However, the combined effect of METH and HIV-1 Tat on neurocognition and its potential effect on neuroplasticity mechanisms remains largely unknown. Therefore, the present study was undertaken to investigate the combined effect of METH and HIV-1 Tat on behavior and on the expression of neuroplasticity markers by utilizing Doxycycline (DOX)-inducible HIV-1 Tat (1-86) transgenic mice. Expression of Tat in various brain regions of these mice was confirmed by RT-PCR. The mice were administered with an escalating dose of METH (0.1 mg/kg to 6 mg/kg, i.p) over a 7-day period, followed by 6 mg/kg, i.p METH twice a day for four weeks. After three weeks of METH administration, Y maze and Morris water maze assays were performed to determine the effect of Tat and METH on working and spatial memory, respectively. Compared with controls, working memory was significantly decreased in Tat mice that were administered METH. Moreover, significant deficits in spatial memory were also observed in Tat-Tg mice that were administered METH. A significant reduction in the protein expressions of synapsin 1, synaptophysin, Arg3.1, PSD-95, and BDNF in different brain regions were also observed. Expression levels of Calmodulin kinase II (CaMKII), a marker of synaptodendritic integrity, were also significantly decreased in HIV-1 Tat mice that were treated with METH. Together, this data suggests that METH enhances HIV-1 Tat-induced memory deficits by reducing the expression of pre- and postsynaptic proteins and neuroplasticity markers, thus providing novel insights into the molecular mechanisms behind neurocognitive impairments in HIV-infected amphetamine users.


Asunto(s)
Trastornos de la Memoria/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/efectos de los fármacos , Estimulantes del Sistema Nervioso Central , Femenino , Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/metabolismo , Seropositividad para VIH , VIH-1/metabolismo , Humanos , Masculino , Trastornos de la Memoria/metabolismo , Metanfetamina/efectos adversos , Metanfetamina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Crecimiento Nervioso/efectos de los fármacos , Neuronas/metabolismo , Sinapsis/efectos de los fármacos , Sinapsinas/efectos de los fármacos , Sinapsinas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/efectos adversos
7.
J Virol ; 92(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29093081

RESUMEN

Virus-encoded proteins that impair or shut down specific host cell functions during replication can be used as probes to identify potential proteins/pathways used in the replication of viruses from other families. We screened nine proteins from herpes simplex virus 1 (HSV-1) for the ability to enhance or restrict human immunodeficiency virus type 1 (HIV-1) replication. We show that several HSV-1 proteins (glycoprotein M [gM], US3, and UL24) potently restricted the replication of HIV-1. Unlike UL24 and US3, which reduced viral protein synthesis, we observed that gM restriction of HIV-1 occurred through interference with the processing and transport of gp160, resulting in a significantly reduced level of mature gp120/gp41 released from cells. Finally, we show that an HSV-1 gM mutant lacking the majority of the C-terminal domain (HA-gM[Δ345-473]) restricted neither gp160 processing nor the release of infectious virus. These studies identify proteins from heterologous viruses that can restrict viruses through novel pathways.IMPORTANCE HIV-1 infection of humans results in AIDS, characterized by the loss of CD4+ T cells and increased susceptibility to opportunistic infections. Both HIV-1 and HSV-1 can infect astrocytes and microglia of the central nervous system (CNS). Thus, the identification of HSV-1 proteins that directly restrict HIV-1 or interfere with pathways required for HIV-1 replication could lead to novel antiretroviral strategies. The results of this study show that select viral proteins from HSV-1 can potently restrict HIV-1. Further, our results indicate that the gM protein of HSV-1 restricts HIV-1 through a novel pathway by interfering with the processing of gp160 and its incorporation into virus maturing from the cell.


Asunto(s)
VIH-1/fisiología , Herpesvirus Humano 1/fisiología , Interacciones Microbianas , Proteínas Virales/metabolismo , Replicación Viral , Línea Celular , Glicoproteínas/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Proteínas gp160 de Envoltorio del VIH/metabolismo , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteolisis , Proteínas de la Matriz Viral/metabolismo
8.
Virology ; 498: 149-163, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27584592

RESUMEN

Old World monkey (OWM) and hominid APOBEC3Aproteins exhibit differential restriction activities against lentiviruses and DNA viruses. Human APOBEC3A(hA3A)has weak restriction activity against HIV-1Δvifbut is efficiently restricted by an artificially generated chimeric from mandrills (mndA3A/G). We show that a chimeric hA3Acontaining the "WVS" insertion (hA3A[(27)WVS(29)]) conferred potent HIV-1restriction activity. Analysis of each amino acid of the "WVS" motif show that the length and not necessarily the charge or hydrophobicity of the amino acids accounted for restriction activity. Our results suggest that hA3A[(27)WVS(29)]restricts HIV-1at the level of reverse transcription in target cells. Finally, our results suggest that insertion of "WVS" into hA3Amodestly reduces restriction of adeno-associated virus 2(AAV-2)while insertion of the AC Loop1region of the mndA3A/G into hA3A abolished AAV-2 restriction, strengthening the role of this molecular interface in the functional evolution of primate A3A.


Asunto(s)
Aminoácidos/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Resistencia a la Enfermedad , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Mutagénesis Insercional , Infecciones por Parvoviridae/metabolismo , Infecciones por Parvoviridae/virología , Proteínas/genética , Proteínas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Citidina Desaminasa/química , Dependovirus , VIH-1 , Interacciones Huésped-Patógeno , Humanos , Transporte de Proteínas , Proteínas/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Proteínas Virales/metabolismo , Replicación Viral
9.
J Gen Virol ; 97(1): 1-17, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26489798

RESUMEN

The apolipoprotein mRNA editing enzyme catalytic polypeptide-like 3 (APOBEC3; A3) proteins are a family of seven cytidine deaminases (A3A, A3B, A3C, A3D, A3F, A3G and A3H) that restrict certain viral infections. These innate defence factors are best known for their ability to restrict the replication of human immunodeficiency virus type 1 (HIV-1) lacking a functional Vif protein (HIV-1Δvif) through the deamination of cytidine residues to uridines during reverse transcription, ultimately leading to lethal G → A changes in the viral genome. The best studied of the A3 proteins has been APOBEC3G because of its potent activity against HIV-1Δvif. However, one member of this family, A3A, has biological properties that make it unique among the A3 proteins. In this review, we will focus on the structural and phylogenetic features of the human and non-human primate A3A proteins, their role in the restriction of retroviruses and other viruses, and current findings on other biological properties affected by this protein.


Asunto(s)
Citidina Desaminasa/metabolismo , Daño del ADN , Neoplasias/patología , Recombinación Genética , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Humanos , Primates , Infecciones por Retroviridae/virología
10.
Immunogenetics ; 67(7): 355-70, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25994147

RESUMEN

B cell functional defects are associated with delayed neutralizing antibody development in pathogenic lentivirus infections. However, the timeframe for alterations in the antibody repertoire and somatic hypermutation (SHM) remains unclear. Here, we utilized the SIV/rhesus macaque (RM) model to investigate the dynamics of immunoglobulin V(H) gene diversity and SHM following infection. Three RMs were infected with SIVmac239, and V(H)1, V(H)3, and V(H)4 genes were amplified from peripheral blood at 0, 2, 6, 24, and 36 weeks postinfection for next-generation sequencing. Analysis of over 3.8 million sequences against currently available RM germline V(H) genes revealed a highly biased V(H) gene repertoire in outbred RMs. SIV infection did not significantly perturb the predominant IgG1 response, but overall immunoglobulin SHM declined during the course of SIV infection. Moreover, SHM at the AID deamination hotspot, WRC, rapidly decreased and was suppressed throughout SIV infection. In contrast, a transient increase in mutations at the APOBEC3G deamination hotspot, CCC, coincided with a spike in APOBEC3G expression during acute SIV infection. The results outline a timetable for altered V(H) gene repertoire and IgG SHM in the SIV/RM model and suggest a burst of APOBEC3G-mediated antibody SHM during acute SIV infection.


Asunto(s)
Linfocitos B/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Hipermutación Somática de Inmunoglobulina/genética , Animales , Diversidad de Anticuerpos , Secuencia de Bases , Citidina Desaminasa/genética , Frecuencia de los Genes , Variación Genética/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Macaca mulatta , Análisis de Secuencia de ADN , Virus de la Inmunodeficiencia de los Simios/inmunología
11.
J Immunol ; 193(11): 5613-25, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25362178

RESUMEN

We previously demonstrated that HIV replication is concentrated in lymph node B cell follicles during chronic infection and that HIV-specific CTL fail to accumulate in large numbers at those sites. It is unknown whether these observations can be generalized to other secondary lymphoid tissues or whether virus compartmentalization occurs in the absence of CTL. We evaluated these questions in SIVmac239-infected rhesus macaques by quantifying SIV RNA(+) cells and SIV-specific CTL in situ in spleen, lymph nodes, and intestinal tissues obtained at several stages of infection. During chronic asymptomatic infection prior to simian AIDS, SIV-producing cells were more concentrated in follicular (F) compared with extrafollicular (EF) regions of secondary lymphoid tissues. At day 14 of infection, when CTL have minimal impact on virus replication, there was no compartmentalization of SIV-producing cells. Virus compartmentalization was diminished in animals with simian AIDS, which often have low-frequency CTL responses. SIV-specific CTL were consistently more concentrated within EF regions of lymph node and spleen in chronically infected animals regardless of epitope specificity. Frequencies of SIV-specific CTL within F and EF compartments predicted SIV RNA(+) cells within these compartments in a mixed model. Few SIV-specific CTL expressed the F homing molecule CXCR5 in the absence of the EF retention molecule CCR7, possibly accounting for the paucity of F CTL. These findings bolster the hypothesis that B cell follicles are immune privileged sites and suggest that strategies to augment CTL in B cell follicles could lead to improved viral control and possibly a functional cure for HIV infection.


Asunto(s)
Ganglios Linfáticos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos Virales/inmunología , Movimiento Celular , Células Cultivadas , Progresión de la Enfermedad , Macaca mulatta , ARN Viral/análisis , Receptores CCR7/metabolismo , Receptores CXCR5/metabolismo , Linfocitos T Citotóxicos/virología , Replicación Viral
12.
Virology ; 468-470: 532-544, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25262471

RESUMEN

The APOBEC3 (A3) deaminases are retrovirus restriction factors that were proposed as inhibitory components of HIV-1 gene therapy vectors. However, A3 mutational activity may induce undesired genomic damage and enable HIV-1 to evade drugs and immune responses. Here, we show that A3A protein from Colobus guereza (colA3A) can restrict HIV-1 replication in producer cells in a deaminase-independent manner without inducing DNA damage. Neither HIV-1 reverse transcription nor integration were significantly affected by colA3A, but capsid protein synthesis was inhibited. The determinants for colA3A restriction mapped to the N-terminal region. These properties extend to A3A from mandrills and De Brazza's monkeys. Surprisingly, truncated colA3A proteins expressing only the N-terminal 100 amino acids effectively exclude critical catalytic regions but retained potent cellular restriction activity. These highlight a unique mechanism of cellular HIV-1 restriction by several Old World monkey A3A proteins that may be exploited for functional HIV-1 cure strategies.


Asunto(s)
Colobus , Citidina Desaminasa/metabolismo , VIH-1/genética , VIH-1/fisiología , Replicación Viral/fisiología , Animales , Línea Celular , Citidina Desaminasa/genética , Virus de la Leucemia Murina de Friend/fisiología , Regulación Enzimológica de la Expresión Génica , Humanos , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes
13.
Virology ; 449: 140-9, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24418547

RESUMEN

Previous studies have shown that apolipoprotein B mRNA editing, enzyme catalytic, polypeptide G (APOBEC3G; hA3G) and F (APOBEC3F; hA3F) proteins interact with a nonlinear binding site located at the N-terminal region of the HIV-1 Vif protein. We have analyzed the role of 12 positively charged amino acids of the N-terminal region of the SIV Vif. Simian-human immunodeficiency viruses (SHIV) were constructed that expressed each of these amino acid substitutions. These viruses were examined for replication in the presence of rhesus macaque APOBEC3 proteins (rhA3A-rhA3H), incorporation of the different A3 proteins into virions, and replication in rhesus macaque PBMC. Similar to other studies, we found that K27 was essential for rhA3G activity and rhA3F but was not important for restriction of SHIVΔvif by rhA3A, rhA3D or rhA3H. Our results identified the arginine at position 14 of the SIV Vif as a critical residue for virus restriction by rhA3D, rhA3G and rhA3H.


Asunto(s)
Citidina Desaminasa/metabolismo , Productos del Gen vif/química , Productos del Gen vif/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/enzimología , Virus de la Inmunodeficiencia de los Simios/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Citidina Desaminasa/antagonistas & inhibidores , Citidina Desaminasa/genética , Productos del Gen vif/genética , Infecciones por VIH/enzimología , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , VIH-1/metabolismo , Humanos , Macaca mulatta , Datos de Secuencia Molecular , Unión Proteica , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/química , Virus de la Inmunodeficiencia de los Simios/genética
14.
Virology ; 442(1): 82-96, 2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23648232

RESUMEN

Rhesus macaque APOBEC3A (rhA3A) is capable of restricting both simian-human immunodeficiency virus (SHIVΔvif) and human immunodeficiency virus (HIV-1Δvif) to a greater extent than hA3A. We constructed chimeric A3A proteins to define the domains required for differential lentivirus restriction. Substitution of amino acids 25-33 from rhA3A into hA3A was sufficient to restrict HIVΔvif to levels similar to rhA3A restriction of SHIVΔvif. We tested if differential lentivirus restriction is conserved between A3A from Old World monkey and hominid lineages. A3A from African green monkey restricted SHIVΔvif but not HIV-1Δvif and colobus monkey A3A restricted both wild type and SHIVΔvif and HIV-1Δvif. In contrast, the gibbon ape A3A restricted neither SHIVΔvif nor HIV-1Δvif. Restriction of SHIVΔvif and HIV-1Δvif by New World monkey A3A proteins was not conserved as the A3A from the squirrel monkey but not the northern owl monkey restricted SHIVΔvif. Finally, the colobus A3A protein appears to restrict by a novel post-entry mechanism.


Asunto(s)
Citidina Desaminasa/metabolismo , VIH-1/patogenicidad , Primates/virología , Proteínas/metabolismo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Replicación Viral , Animales , Línea Celular , Chlorocebus aethiops , Citidina Desaminasa/genética , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Lentivirus/patogenicidad , Lentivirus/fisiología , Macaca mulatta/virología , Proteínas/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología
15.
Virology ; 435(2): 395-405, 2013 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-23218949

RESUMEN

Previously, we reported that simian-human immunodeficiency viruses expressing either the lab adapted subtype B (SHIV(KU-1bMC33)) or subtype C (SHIV(SCVpu)) Vpu proteins of human immunodeficiency virus type 1 (HIV-1) had different rates of CD4(+) T cell loss following inoculation into macaques. In this study, we have generated SHIVs that express either the subtype B or subtype C N-terminal (NTD) and transmembrane (TMD) domains and the opposing cytoplasmic domain (SHIV(VpuBC), SHIV(VpuCB)). In culture systems, SHIV(VpuBC) replicated faster than SHIV(VpuCB) while both proteins exhibited similar ability to down-modulate CD4 surface expression. Following inoculation into macaques, SHIV(VpuBC) resulted in rapid CD4(+) T cell loss similar to the parental SHIV(KU-1bMC33), while the rate of CD4(+) T cell loss in those inoculated with SHIV(VpuCB) was intermediate of SHIV(SCVpu) and SHIV(KU-1bMC33). These results emphasize the importance of the Vpu NTD/TMD region in the rate of CD4(+) T cell loss in the pathogenic X4 SHIV/macaque model.


Asunto(s)
Linfocitos T CD4-Positivos/patología , VIH-1/patogenicidad , Proteínas del Virus de la Inmunodeficiencia Humana/química , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo , Síndrome de Inmunodeficiencia Adquirida/inmunología , Síndrome de Inmunodeficiencia Adquirida/patología , Síndrome de Inmunodeficiencia Adquirida/virología , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , VIH-1/clasificación , VIH-1/genética , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Macaca nemestrina , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/clasificación , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética
16.
PLoS One ; 7(12): e51741, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284757

RESUMEN

During human immunodeficiency virus-1 (HIV-1) assembly, the host proteins CD4 (the HIV-1 receptor) and tetherin (an interferon stimulated anti-viral protein) both reduce viral fitness. The HIV-1 accessory gene Vpu counteracts both of these proteins, but it is thought to do so through two distinct mechanisms. Modulation of CD4 likely occurs through proteasomal degradation from the endoplasmic reticulum. The exact mechanism of tetherin modulation is less clear, with possible roles for degradation and alteration of protein transport to the plasma membrane. Most investigations of Vpu function have used different assays for CD4 and tetherin. In addition, many of these investigations used exogenously expressed Vpu, which could result in variable expression levels. Thus, few studies have investigated these two Vpu functions in parallel assays, making direct comparisons difficult. Here, we present results from a rapid assay used to simultaneously investigate Vpu-targeting of both tetherin and a viral glycoprotein, gibbon ape leukemia virus envelope (GaLV Env). We previously reported that Vpu modulates GaLV Env and prevents its incorporation into HIV-1 particles through a recognition motif similar to that found in CD4. Using this assay, we performed a comprehensive mutagenic scan of Vpu in its native proviral context to identify features required for both types of activity. We observed considerable overlap in the Vpu sequences required to modulate tetherin and GaLV Env. We found that features in the cytoplasmic tail of Vpu, specifically within the cytoplasmic tail hinge region, were required for modulation of both tetherin and GaLV Env. Interestingly, these same regions features have been determined to be critical for CD4 downmodulation. We also observed a role for the transmembrane domain in the restriction of tetherin, as previously reported, but not of GaLV Env. We propose that Vpu may target both proteins in a mechanistically similar manner, albeit in different cellular locations.


Asunto(s)
Antígenos CD/metabolismo , VIH-1/crecimiento & desarrollo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Virus de la Leucemia del Gibón/crecimiento & desarrollo , Proteínas Mutantes/metabolismo , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Secuencia de Aminoácidos , Antígenos CD/genética , Citoplasma/metabolismo , Citometría de Flujo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HEK293/metabolismo , Células HEK293/virología , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Virus de la Leucemia del Gibón/genética , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Reguladoras y Accesorias Virales/genética , Virión/metabolismo , Ensamble de Virus , Replicación Viral
17.
J Biol Chem ; 287(1): 58-67, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22072710

RESUMEN

The Vpu protein of HIV-1 antagonizes BST-2 (tetherin), a broad spectrum effector of the innate immune response to viral infection, by an intermolecular interaction that maps genetically to the α-helical transmembrane domains (TMDs) of each protein. Here we utilize NMR spectroscopy to describe key features of the helix-helix pairing that underlies this interaction. The antagonism of BST-2 involves a sequence of three alanines and a tryptophan spaced at four residue intervals within the Vpu TMD helix. Responsiveness to Vpu involves bulky hydrophobic residues in the C-terminal region of the BST-2 TMD helix that likely fit between the alanines on the interactive face of Vpu. These aspects of Vpu and BST-2 form an anti-parallel, lipid-embedded helix-helix interface. Changes in human BST-2 that mimic sequences found in nonhuman primate orthologs unresponsive to Vpu change the tilt angle of the TMD in the lipid bilayer without abrogating its intrinsic ability to interact with Vpu. These data explain the mechanism by which HIV-1 evades a key aspect of innate immunity and the species specificity of Vpu using an anti-parallel helix-helix packing model.


Asunto(s)
Antígenos CD/química , Antígenos CD/metabolismo , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/química , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Inmunidad Innata , Membrana Dobles de Lípidos/metabolismo , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo , Secuencia de Aminoácidos , Antígenos CD/inmunología , Membrana Celular/metabolismo , Membrana Celular/virología , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , VIH-1/inmunología , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/química , Micelas , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Termodinámica
18.
Virology ; 419(1): 24-42, 2011 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-21868050

RESUMEN

The human apolipoprotein B mRNA editing enzyme catalytic peptide-like 3 (APOBEC3; A3) family of proteins (A3A-H) are known to restrict various retroviruses and retroelements, but the full complement of rhesus macaque A3 proteins remains unclear. We report the isolation and characterization of the hA3A homologue from rhesus macaques (rhA3A) and show that the rhesus macaque and human A3 genes are orthologous. RhA3A is expressed at high levels in activated CD4+ T cells, is widely expressed in macaque tissues, and is degraded in the presence of the human immunodeficiency virus (HIV-1) and simian-human immunodeficiency virus (SHIV) genomes. Our results indicate that rhA3A is a potent inhibitor of SHIVΔvif and to a lesser extent HIV-1Δvif. Unlike hA3A, rhA3A did not inhibit adeno-associated virus 2 (AAV-2) replication and L1 retrotransposition. These data suggest an evolutionary switch in primate A3A virus specificity and provide the first evidence that a primate A3A can inhibit lentivirus replication.


Asunto(s)
Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Evolución Molecular , Macaca mulatta/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Desaminasas APOBEC-1 , Animales , Linfocitos T CD4-Positivos/enzimología , Dependovirus/inmunología , Perfilación de la Expresión Génica , VIH/inmunología , Humanos , Datos de Secuencia Molecular , Retroelementos/inmunología , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
19.
Virology ; 408(1): 89-102, 2010 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-20880565

RESUMEN

The Vpu protein of human immunodeficiency virus type 1 (HIV-1) is known to enhance virion release from certain cell types. To accomplish this function, Vpu interacts with the restriction factor known as bone marrow stromal cell antigen 2 (BST-2)/tetherin. In this study, we analyzed whether the Vpu protein is associated with microdomains known as lipid or membrane rafts. Our results indicate that Vpu partially partitions into detergent-resistant membrane (DRM) fractions when expressed alone or in the context of simian-human immunodeficiency virus (SHIV) infection. The ability to be partitioned into rafts was observed with both subtype B and C Vpu proteins. The use of cholesterol lowering lovastatin/M-ß-cyclodextrin and co-patching experiments confirmed that Vpu can be detected in cholesterol rich regions of membranes. Finally, we present data showing that raft association-defective transmembrane mutants of Vpu have impaired enhanced virus release function, but still maintain the ability to down-regulate CD4.


Asunto(s)
VIH-1/fisiología , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Microdominios de Membrana/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Liberación del Virus , Anticolesterolemiantes/farmacología , Línea Celular , Colesterol/metabolismo , Humanos , Lovastatina/farmacología
20.
Virology ; 406(2): 312-21, 2010 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-20708210

RESUMEN

Pathogenic simian-human immunodeficiency viruses (SHIV) contain HIV-1 Vpu and SIV Nef, both shown to counteract BST-2 (HM1.24; CD317; tetherin) inhibition of virus release in a species-specific manner. We show that human and pig-tailed BST-2 (ptBST-2) restrict SHIV. We found that sequential "humanization" of the transmembrane domain (TMD) of the pig-tailed BST-2 (ptBST-2) protein resulted in a fluctuation in sensitivity to HIV-1 Vpu. Our results also show that the length of the TMD in human and ptBST-2 proteins is important for BST-2 restriction and susceptibility to Vpu. Taken together, our results emphasize the importance of tertiary structure in BST-2 antagonism and suggests that the HIV-1 Vpu transmembrane domain may have additional functions in vivo unrelated to BST-2 antagonism.


Asunto(s)
Antígenos CD/metabolismo , Infecciones por VIH/metabolismo , VIH-1/fisiología , Glicoproteínas de Membrana/metabolismo , Virus Reordenados/fisiología , Virus de la Inmunodeficiencia de los Simios/fisiología , Secuencia de Aminoácidos , Animales , Antígenos CD/química , Antígenos CD/genética , Línea Celular , Proteínas Ligadas a GPI , Productos del Gen nef/genética , Productos del Gen nef/metabolismo , Infecciones por VIH/virología , VIH-1/genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Macaca mulatta , Macaca nemestrina , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Virus Reordenados/genética , Alineación de Secuencia , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA