Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Retrovirology ; 8: 9, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21310048

RESUMEN

BACKGROUND: The cellular transmembrane protein CD317/BST-2/HM1.24/Tetherin restricts HIV-1 infection by physically tethering mature virions to the surface of infected cells. HIV-1 counteracts this restriction by expressing the accessory protein Vpu, yet the mechanism of this antagonism is incompletely understood. ß-TrCP is the substrate recognition domain of an E3 ubiquitin ligase complex that interacts with the di-serine motif S52/S56 in the cytoplasmic tail of Vpu to target the CD4 receptor for proteasomal degradation. Recently, it has been suggested that ß-TrCP is also critically involved in Vpu's ability to overcome the CD317-mediated virion release block. RESULTS: To test this model, we analyzed the consequences of several experimental strategies to interfere with the Vpu-ß-TrCP protein-protein interaction. Under these conditions, we studied effects of Vpu on expression and localization of CD317 and CD4, as well as on its ability to promote HIV-1 release. Our results demonstrate a strict requirement for Vpu's di-serine motif for degradation of CD4 and also CD317, reduction of cell surface exposure of CD317, and HIV-1 release enhancement. We further show a critical role of ß-TrCP2, but not of the structurally related ß-TrCP1 isoform, for Vpu-mediated degradation of both receptors. Most importantly, Vpu remained active in downregulating CD317 from the cell surface and in overcoming the HIV-1 release restriction in ß-TrCP-depleted cells. CONCLUSIONS: These results demonstrate that ß-TrCP is not strictly required for Vpu's ability to counteract the CD317-imposed virion release block and support the relevance of cell surface down-modulation of the restriction factor as a central mechanism of Vpu antagonism. Moreover, we propose the existence of a critical, yet to be identified cellular factor that interacts with Vpu via its di-serine motif to alter the trafficking of the restriction factor.


Asunto(s)
Antígenos CD/metabolismo , VIH-1/fisiología , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Antígenos CD/genética , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/virología , Regulación hacia Abajo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Proteínas Reguladoras y Accesorias Virales/genética , Virión/metabolismo , Virión/fisiología , Liberación del Virus/fisiología , Proteínas con Repetición de beta-Transducina/genética
2.
J Virol ; 84(23): 12300-14, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20861260

RESUMEN

An immunocompetent, permissive, small-animal model would be valuable for the study of human immunodeficiency virus type 1 (HIV-1) pathogenesis and for the testing of drug and vaccine candidates. However, the development of such a model has been hampered by the inability of primary rodent cells to efficiently support several steps of the HIV-1 replication cycle. Although transgenesis of the HIV receptor complex and human cyclin T1 have been beneficial, additional late-phase blocks prevent robust replication of HIV-1 in rodents and limit the range of in vivo applications. In this study, we explored the HIV-1 susceptibility of rabbit primary T cells and macrophages. Envelope-specific and coreceptor-dependent entry of HIV-1 was achieved by expressing human CD4 and CCR5. A block of HIV-1 DNA synthesis, likely mediated by TRIM5, was overcome by limited changes to the HIV-1 gag gene. Unlike with mice and rats, primary cells from rabbits supported the functions of the regulatory viral proteins Tat and Rev, Gag processing, and the release of HIV-1 particles at levels comparable to those in human cells. While HIV-1 produced by rabbit T cells was highly infectious, a macrophage-specific infectivity defect became manifest by a complex pattern of mutations in the viral genome, only part of which were deamination dependent. These results demonstrate a considerable natural HIV-1 permissivity of the rabbit species and suggest that receptor complex transgenesis combined with modifications in gag and possibly vif of HIV-1 to evade species-specific restriction factors might render lagomorphs fully permissive to infection by this pathogenic human lentivirus.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por VIH/transmisión , VIH-1/genética , Macrófagos/virología , Linfocitos T/virología , Internalización del Virus , Replicación Viral/fisiología , Animales , Análisis Mutacional de ADN , ADN Complementario/genética , Duplicado del Terminal Largo de VIH/genética , Células HeLa , Humanos , Immunoblotting , Ratones , Células 3T3 NIH , Conejos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
J Biol Chem ; 284(43): 29692-703, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19666477

RESUMEN

Protease inhibitors (PI) act by blocking human immunodeficiency virus (HIV) polyprotein processing, but there is no direct quantitative correlation between the degree of impairment of Gag processing and virion infectivity at low PI concentrations. To analyze the consequences of partial processing, virus particles were produced in the presence of limiting PI concentrations or by co-transfection of wild-type proviral plasmids with constructs carrying mutations in one or more cleavage sites. Low PI concentrations caused subtle changes in polyprotein processing associated with a pronounced reduction of particle infectivity. Dissection of individual stages of viral entry indicated a block in accumulation of reverse transcriptase products, whereas virus entry, enzymatic reverse transcriptase activity, and replication steps following reverse transcription were not affected. Co-expression of low amounts of partially processed forms of Gag together with wild-type HIV generally exerted a trans-dominant effect, which was most prominent for a construct carrying mutations at both cleavage sites flanking the CA domain. Interestingly, co-expression of low amounts of Gag mutated at the CA-SP1 cleavage site also affected processing activity at this site in the wild-type virus. The results indicate that low amounts (<5%) of Gag processing intermediates can display a trans-dominant effect on HIV particle maturation, with the maturation cleavage between CA and SP1 being of particular importance. These effects are likely to be important for the strong activity of PI at concentrations achieved in vivo and also bear relevance for the mechanism of action of the antiviral drug bevirimat.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacología , VIH-1/patogenicidad , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Succinatos/farmacología , Triterpenos/farmacología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Transcriptasa Inversa del VIH/genética , Transcriptasa Inversa del VIH/metabolismo , VIH-1/fisiología , Células HeLa , Humanos , Mutación , Estructura Terciaria de Proteína/genética , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
4.
J Virol ; 83(17): 8869-84, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19535440

RESUMEN

Latent cytomegalovirus (CMV) is frequently transmitted by organ transplantation, and its reactivation under conditions of immunosuppressive prophylaxis against graft rejection by host-versus-graft disease bears a risk of graft failure due to viral pathogenesis. CMV is the most common cause of infection following liver transplantation. Although hematopoietic cells of the myeloid lineage are a recognized source of latent CMV, the cellular sites of latency in the liver are not comprehensively typed. Here we have used the BALB/c mouse model of murine CMV infection to identify latently infected hepatic cell types. We performed sex-mismatched bone marrow transplantation with male donors and female recipients to generate latently infected sex chromosome chimeras, allowing us to distinguish between Y-chromosome (gene sry or tdy)-positive donor-derived hematopoietic descendants and Y-chromosome-negative cells of recipients' tissues. The viral genome was found to localize primarily to sry-negative CD11b(-) CD11c(-) CD31(+) CD146(+) cells lacking major histocompatibility complex class II antigen (MHC-II) but expressing murine L-SIGN. This cell surface phenotype is typical of liver sinusoidal endothelial cells (LSECs). Notably, sry-positive CD146(+) cells were distinguished by the expression of MHC-II and did not harbor latent viral DNA. In this model, the frequency of latently infected cells was found to be 1 to 2 per 10(4) LSECs, with an average copy number of 9 (range, 4 to 17) viral genomes. Ex vivo-isolated, latently infected LSECs expressed the viral genes m123/ie1 and M122/ie3 but not M112-M113/e1, M55/gB, or M86/MCP. Importantly, in an LSEC transfer model, infectious virus reactivated from recipients' tissue explants with an incidence of one reactivation per 1,000 viral-genome-carrying LSECs. These findings identified LSECs as the main cellular site of murine CMV latency and reactivation in the liver.


Asunto(s)
Células Endoteliales/virología , Hígado/virología , Muromegalovirus/fisiología , Activación Viral , Latencia del Virus , Animales , Femenino , Perfilación de la Expresión Génica , Genes Virales , Masculino , Ratones , Ratones Endogámicos BALB C
5.
Retrovirology ; 5: 58, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18611257

RESUMEN

BACKGROUND: The development of an immunocompetent, genetically modified mouse model to study HIV-1 pathogenesis and to test antiviral strategies has been hampered by the fact that cells from native mice do not or only inefficiently support several steps of the HIV-1 replication cycle. Upon HIV-1 infection, mouse T-cell lines fail to express viral proteins, but the underlying replication barrier has thus far not been unambiguously identified. Here, we performed a kinetic and quantitative assessment of consecutive steps in the early phase of the HIV-1 replication cycle in T-cells from mice and humans. RESULTS: Both T-cell lines and primary T-cells from mice harbor a severe post-entry defect that is independent of potential species-specTR transactivation. Reverse transcription occurred efficiently following VSV-G-mediated entry of virions into mouse T-cells, and abundant levels of 2-LTR circles indicated successful nuclear import of the pre-integration complex. To probe the next step in the retroviral replication cycle, i.e. the integration of HIV-1 into the host cell genome, we established and validated a nested real-time PCR to specifically quantify HIV-1 integrants exploiting highly repetitive mouse B1 elements. Importantly, we demonstrate that the frequency of integrant formation is diminished 18- to > 305-fold in mouse T-cell lines compared to a human counterpart, resulting in a largely abortive infection. Moreover, differences in transgene expression from residual vector integrants, the transcription off which is cyclin T1-independent, provided evidence for an additional, peri-integrational deficit in certain mouse T-cell lines. CONCLUSION: In contrast to earlier reports, we find that mouse T-cells efficiently support early replication steps up to and including nuclear import, but restrict HIV-1 at the level of chromosomal integration.


Asunto(s)
VIH-1/fisiología , Linfocitos T/virología , Integración Viral , Animales , Línea Celular , ADN Viral/biosíntesis , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Transcripción Reversa , Transcripción Genética , Internalización del Virus
6.
J Immunol Methods ; 336(1): 85-9, 2008 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-18471825

RESUMEN

Efficient gene delivery is essential for the assessment of transgene function in untransformed hematopoietic cells. Here, we explored the utility of different non-viral and viral gene delivery techniques for primary T cells from New Zealand White rabbits. We find that electroporation and nucleofection result in high-level transgene expression from both small and large GFP reporter constructs in activated rabbit T cells at moderate cytotoxicity. Both non-viral gene delivery methods were vastly superior to retroviral, lentiviral, or adenoviral transduction approaches. The effectiveness of non-viral gene delivery for functional analyses was demonstrated by downregulation of CD4 cell surface molecules through transient expression of the endocytosis-inducing Nef protein from human immunodeficiency virus in a signature motif-specific manner. This study establishes conventional electroporation as an efficient and inexpensive procedure to render primary rabbit T cells accessible to rapid functional ex vivo analyses.


Asunto(s)
Conejos/fisiología , Linfocitos T/inmunología , Transfección/métodos , Animales , Animales Modificados Genéticamente , Antígenos CD4/genética , Antígenos CD4/inmunología , Supervivencia Celular/inmunología , Electroporación/métodos , Femenino , Citometría de Flujo/veterinaria , Formazáns/química , Proteínas Fluorescentes Verdes/genética , Modelos Animales , Sales de Tetrazolio/química , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología
7.
Retrovirology ; 4: 53, 2007 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-17655755

RESUMEN

BACKGROUND: In vivo studies on HIV-1 pathogenesis and testing of antiviral strategies have been hampered by the lack of an immunocompetent small animal model that is highly susceptible to HIV-1 infection. Since native rodents are non-permissive, we developed transgenic rats that selectively express the HIV-1 receptor complex, hCD4 and hCCR5, on relevant target cells. These animals display a transient low-level plasma viremia after HIV-1YU-2 infection, demonstrating HIV-1 susceptibility in vivo. However, unlike macrophages, primary CD4 T-cells from double-transgenic animals fail to support viral spread ex vivo. To identify quantitative limitations or absolute blocks in this rodent species, we quantitatively assessed the efficiency of key steps in the early phase of the viral replication cycle in a side-by-side comparison in infected cell lines and primary T-cells from hCD4/hCCR5-transgenic rats and human donors. RESULTS: Levels of virus entry, HIV-1 cDNA synthesis, nuclear import, and integration into the host genome were shown to be remarkably similar in cell lines and, where technically accessible, in primary T-cells from both species. In contrast, a profound impairment at the level of early HIV gene expression was disclosed at the single-cell level in primary rat T-cells and most other rat-derived cells. Macrophages were a notable exception, possibly reflecting the unique transcriptional milieu in this evolutionarily conserved target cell of all lentiviruses. Importantly, transient trans-complementation by ex vivo nucleofection with the Tat-interacting protein Cyclin T1 of human origin markedly elevated HIV gene expression in primary rat T-cells. CONCLUSION: This is the first study that has quantitatively determined the efficiency of consecutive steps in the HIV-1 replication cycle in infected primary HIV target cells from a candidate transgenic small animal and compared it to human cells. Unlike cells derived from mice or rabbits, rat cells complete all of the early steps in the HIV-1 replication cycle, including provirus integration in vivo, with high efficiency. A deficiency in gene expression was disclosed at the single cell level and could be counteracted by the human pTEFb transcription complex factor Cyclin T1. Collectively, these results provide the basis for the advancement of this transgenic rat model through strategies aimed at boosting HIV-1 gene expression in primary rat CD4 T-cells, including human Cyclin T1 transgenesis.


Asunto(s)
Antígenos CD4/metabolismo , Modelos Animales de Enfermedad , Regulación Viral de la Expresión Génica , Infecciones por VIH/virología , VIH-1/fisiología , Receptores CCR5/metabolismo , Linfocitos T/virología , Replicación Viral , Animales , Animales Modificados Genéticamente , Antígenos CD4/genética , Células Cultivadas , Ciclina T , Ciclinas/metabolismo , Femenino , Infecciones por VIH/inmunología , Infecciones por VIH/fisiopatología , VIH-1/patogenicidad , Células HeLa , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratas , Receptores CCR5/genética , Internalización del Virus
8.
Cell Host Microbe ; 5(3): 285-97, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19286137

RESUMEN

Mammals encode proteins that inhibit viral replication at the cellular level. In turn, certain viruses have evolved genes that can functionally counteract these intrinsic restrictions. Human CD317 (BST-2/HM1.24/tetherin) is a restriction factor that blocks release of human immunodeficiency virus type 1 (HIV-1) from the cell surface and can be overcome by HIV-1 Vpu. Here, we show that mouse and rat CD317 potently inhibit HIV-1 release but are resistant to Vpu. Interspecies chimeras reveal that the rodent-specific resistance and human-specific sensitivity to Vpu antagonism involve all three major structural domains of CD317. To promote virus release, Vpu depletes cellular pools of human CD317, but not of the rodent orthologs, by accelerating its degradation via the 20S proteasome. Thus, HIV-1 Vpu suppresses the expression of the CD317 antiviral factor in human cells, and the species-specific resistance to this suppression may guide the development of small animal models of HIV infection.


Asunto(s)
Antígenos CD/inmunología , VIH-1/inmunología , VIH-1/patogenicidad , Proteínas del Virus de la Inmunodeficiencia Humana/fisiología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/inmunología , Proteínas Reguladoras y Accesorias Virales/fisiología , Factores de Virulencia/fisiología , Animales , Antígenos CD/metabolismo , Línea Celular , Proteínas Ligadas a GPI , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratas
9.
J Virol ; 80(21): 10436-56, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16928768

RESUMEN

During murine cytomegalovirus (mCMV) latency in the lungs, most of the viral genomes are transcriptionally silent at the major immediate-early locus, but rare and stochastic episodes of desilencing lead to the expression of IE1 transcripts. This low-frequency but perpetual expression is accompanied by an activation of lung-resident effector-memory CD8 T cells specific for the antigenic peptide 168-YPHFMPTNL-176, which is derived from the IE1 protein. These molecular and immunological findings were combined in the "silencing/desilencing and immune sensing hypothesis" of cytomegalovirus latency and reactivation. This hypothesis proposes that IE1 gene expression proceeds to cell surface presentation of the IE1 peptide by the major histocompatibility complex (MHC) class I molecule L(d) and that its recognition by CD8 T cells terminates virus reactivation. Here we provide experimental evidence in support of this hypothesis. We generated mutant virus mCMV-IE1-L176A, in which the antigenic IE1 peptide is functionally deleted by a point mutation of the C-terminal MHC class I anchor residue Leu into Ala. Two revertant viruses, mCMV-IE1-A176L and the wobble nucleotide-marked mCMV-IE1-A176L*, in which Leu is restored by back-mutation of Ala codon GCA into Leu codons CTA and CTT, respectively, were constructed. Pulmonary latency of the mutant virus was found to be associated with an increased prevalence of IE1 transcription and with events of IE3 transactivator splicing. In conclusion, IE1-specific CD8 T cells recognize and terminate virus reactivation in vivo at the first opportunity in the reactivated gene expression program. The perpetual gene expression and antigen presentation might represent the driving molecular force in CMV-associated immunosenescence.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Pulmón/inmunología , Pulmón/virología , Muromegalovirus/inmunología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Antígenos Virales/genética , Secuencia de Bases , Trasplante de Médula Ósea , ADN Viral/genética , Epítopos/genética , Femenino , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Huésped Inmunocomprometido , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Muromegalovirus/genética , Muromegalovirus/patogenicidad , Muromegalovirus/fisiología , Mutagénesis Sitio-Dirigida , Fenotipo , Transactivadores/genética , Transactivadores/inmunología , Activación Transcripcional , Latencia del Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA