Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell ; 146(6): 918-30, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925315

RESUMEN

Inhibitors of DNA binding (IDs) antagonize basic-helix-loop-helix (bHLH) transcription factors to inhibit differentiation and maintain stem cell fate. ID ubiquitination and proteasomal degradation occur in differentiated tissues, but IDs in many neoplasms appear to escape degradation. We show that the deubiquitinating enzyme USP1 promotes ID protein stability and stem cell-like characteristics in osteosarcoma. USP1 bound, deubiquitinated, and thereby stabilized ID1, ID2, and ID3. A subset of primary human osteosarcomas coordinately overexpressed USP1 and ID proteins. USP1 knockdown in osteosarcoma cells precipitated ID protein destabilization, cell-cycle arrest, and osteogenic differentiation. Conversely, ectopic USP1 expression in mesenchymal stem cells stabilized ID proteins, inhibited osteoblastic differentiation, and enhanced proliferation. Consistent with USP1 functioning in normal mesenchymal stem cells, USP1-deficient mice were osteopenic. Our observations implicate USP1 in preservation of the stem cell state that characterizes osteosarcoma and identify USP1 as a target for differentiation therapy.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Neoplásicas/citología , Osteosarcoma/patología , Animales , Proteínas de Arabidopsis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteasas Ubiquitina-Específicas , Ubiquitinación
2.
Lab Invest ; 93(9): 970-82, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23917877

RESUMEN

Cancer is a heterogeneous disease manifest in many forms. Tumor histopathology can differ significantly among patients and cellular heterogeneity within tumors is common. A primary goal of cancer biologists is to better understand tumorigenesis and cancer progression; however, the complex nature of tumors has posed a substantial challenge to unlocking cancer's secrets. The cancer stem cell (CSC) paradigm for the pathobiology of solid tumors appropriately acknowledges phenotypic and functional tumor cell heterogeneity observed in solid tumors and accounts for the disconnect between drug approval based on response and the general inability of approved therapies to meaningfully impact survival due to their failure to eradicate these most important of cellular targets. First proposed to exist decades ago, CSC have only recently begun to be precisely identified due to technical advancements that facilitate identification, isolation, and interrogation of distinct tumor cell subpopulations with differing ability to form and perpetuate tumors. Precise identification of CSC populations and the complete hierarchy of cells within solid tumors will facilitate more accurate characterization of patient subtypes and ultimately contribute to more personalized and effective therapies. Rapid advancement in the understanding of tumor biology as it exists in patients requires cooperation among institutions, surgeons, pathologists, cancer biologists and patients alike, primarily because this translational research is best done with patient-derived tissue grown in the xenograft setting as patient-derived xenografts. This review calls for a broader change in the approaches taken to study cancer pathobiology, highlights what implications the CSC paradigm has for pathologists and cancer biologists alike, and calls for greater collaboration between institutions, physicians and scientists in order to more rapidly advance our collective understanding of cancer.


Asunto(s)
Trasplante de Neoplasias/métodos , Neoplasias/patología , Células Madre Neoplásicas/patología , Trasplante Heterólogo/patología , Animales , Humanos , Oncología Médica
3.
Mol Cell Biol ; 25(18): 7966-75, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16135789

RESUMEN

The nuclear functions of NF-kappaB p50/RelA heterodimers are regulated in part by posttranslational modifications of its RelA subunit, including phosphorylation and acetylation. Acetylation at lysines 218, 221, and 310 differentially regulates RelA's DNA binding activity, assembly with IkappaBalpha, and transcriptional activity. However, it remains unclear whether the acetylation is regulated or simply due to stimulus-coupled nuclear translocation of NF-kappaB. Using anti-acetylated lysine 310 RelA antibodies, we detected p300-mediated acetylation of RelA in vitro and in vivo after stimulation of cells with tumor necrosis factor alpha (TNF-alpha). Coexpression of catalytically inactive mutants of the catalytic subunit of protein kinase A/mitogen- and stress-activated kinase 1 or IKK1/IKK2, which phosphorylate RelA on serine 276 or serine 536, respectively, sharply inhibited RelA acetylation on lysine 310. Furthermore, phosphorylation of RelA on serine 276 or serine 536 increased assembly of phospho-RelA with p300, which enhanced acetylation on lysine 310. Reconstitution of RelA-deficient murine embryonic fibroblasts with RelA S276A or RelA S536A decreased TNF-alpha-induced acetylation of lysine 310 and expression of the endogenous NF-kappaB-responsive E-selectin gene. These findings indicate that the acetylation of RelA at lysine 310 is importantly regulated by prior phosphorylation of serines 276 and 536. Such phosphorylated and acetylated forms of RelA display enhanced transcriptional activity.


Asunto(s)
FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Acetilación , Animales , Células Cultivadas , Proteína p300 Asociada a E1A , Humanos , Lisina/metabolismo , Ratones , Mutación , FN-kappa B/genética , Fosforilación , Serina/metabolismo , Factor de Transcripción ReIA
4.
Trends Microbiol ; 13(4): 137-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15817380

RESUMEN

A recent study has provided important clues towards the identity of the host genes that conspire to promote post-integration latency of human immunodeficiency virus (HIV). Various genes controlling transcription, histone deacetylation and proteasome-mediated protein degradation have emerged as potential players. If the desired, but difficult, goal of complete virus eradication in HIV-infected patients is ever to be realized, the latent reservoir of HIV proviruses must be cleared. Understanding the molecular basis for viral latency is the key first step.


Asunto(s)
Linfocitos T CD4-Positivos/virología , VIH/fisiología , Provirus/fisiología , Integración Viral , Latencia del Virus , Linfocitos T CD4-Positivos/fisiología , Regulación Viral de la Expresión Génica , VIH/genética , Humanos , Provirus/genética , Integración Viral/genética , Latencia del Virus/genética
5.
Sci Transl Med ; 7(302): 302ra136, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26311731

RESUMEN

The high-grade pulmonary neuroendocrine tumors, small cell lung cancer (SCLC) and large cell neuroendocrine carcinoma (LCNEC), remain among the most deadly malignancies. Therapies that effectively target and kill tumor-initiating cells (TICs) in these cancers should translate to improved patient survival. Patient-derived xenograft (PDX) tumors serve as excellent models to study tumor biology and characterize TICs. Increased expression of delta-like 3 (DLL3) was discovered in SCLC and LCNEC PDX tumors and confirmed in primary SCLC and LCNEC tumors. DLL3 protein is expressed on the surface of tumor cells but not in normal adult tissues. A DLL3-targeted antibody-drug conjugate (ADC), SC16LD6.5, comprised of a humanized anti-DLL3 monoclonal antibody conjugated to a DNA-damaging pyrrolobenzodiazepine (PBD) dimer toxin, induced durable tumor regression in vivo across multiple PDX models. Serial transplantation experiments executed with limiting dilutions of cells provided functional evidence confirming that the lack of tumor recurrence after SC16LD6.5 exposure resulted from effective targeting of DLL3-expressing TICs. In vivo efficacy correlated with DLL3 expression, and responses were observed in PDX models initiated from patients with both limited and extensive-stage disease and were independent of their sensitivity to standard-of-care chemotherapy regimens. SC16LD6.5 effectively targets and eradicates DLL3-expressing TICs in SCLC and LCNEC PDX tumors and is a promising first-in-class ADC for the treatment of high-grade pulmonary neuroendocrine tumors.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de la Membrana/inmunología , Tumores Neuroendocrinos/tratamiento farmacológico , Animales , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Tumores Neuroendocrinos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
DNA Cell Biol ; 21(9): 679-88, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12396611

RESUMEN

HIV-1 viral protein R (Vpr) is a small, highly conserved accessory protein encoded by the HIV genome that serves many functions in the viral life cycle. Vpr induces G2 cell cycle arrest, which is thought to indirectly enhance viral replication by increasing transcription from the LTR. Vpr has also been implicated in facilitating infection of nondividing cells, most notably macrophages. Because Vpr is a nucleo-cytoplasmic shuttling protein, its role in enhancing viral replication in macrophages may be mediated through enhanced entry of the HIV preintegration complex through the limiting nuclear pore. Free Vpr is detectable in the serum of patients, and in vitro studies implicate extracellular forms of Vpr as an effector of cellular responses mediated through its ability to transduce through intact cytoplasmic membranes. We review the biologic properties of Vpr, focusing on its mechanism of action, role in HIV replication, and significance for host pathogenesis.


Asunto(s)
Productos del Gen vpr/fisiología , VIH-1/fisiología , Transporte Activo de Núcleo Celular , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/fisiología , Núcleo Celular/virología , Ciclinas/metabolismo , Humanos , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
7.
Cytokine ; 39(1): 63-74, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17643313

RESUMEN

HIV-infected patients harbor approximately 10(5)-10(6) memory CD4 T-cells that contain fully integrated but transcriptionally silent HIV proviruses. While small in number, these latently infected cells form a drug-insensitive reservoir that importantly contributes to the life-long persistence of HIV despite highly effective antiviral therapy. In tissue culture, latent HIV proviruses can be activated when their cellular hosts are exposed to select proinflammatory cytokines or their T-cell receptors are ligated. However, due to a lack of potency and/or dose-limiting toxicity, attempts to purge virus from this latent reservoir in vivo with immune-activating agents, such as anti-CD3 antibodies and IL-2, have failed. A deeper understanding of the molecular underpinnings of HIV latency is clearly required, including determining whether viral latency is actively reinforced by transcriptional repressors, defining which inducible host transcription factors most effectively antagonize latency, and elucidating the role of chromatin in viral latency. Only through such an improved understanding will it be possible to identify combination therapies that might allow complete purging of the latent reservoir and to realize the difficult and elusive goal of complete eradication of HIV in infected patients.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH-1/fisiología , Latencia del Virus , Linfocitos T CD4-Positivos/inmunología , Productos del Gen tat/metabolismo , Humanos , Activación de Linfocitos , ARN Polimerasa II/biosíntesis , Activación Viral
8.
J Virol ; 81(11): 6043-56, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17376917

RESUMEN

Cells harboring infectious, but transcriptionally latent, human immunodeficiency virus type 1 (HIV-1) proviruses currently pose an insurmountable barrier to viral eradication in infected patients. To better understand the molecular basis for HIV-1 latency, we used the J-Lat model of postintegration HIV-1 latency to assess the kinetic relationship between the induction of NF-kappaB and the activation of latent HIV-1 gene expression. Chromatin immunoprecipitation analyses revealed an oscillating pattern of RelA recruitment to the HIV-1 long terminal repeat (LTR) during continuous tumor necrosis factor alpha (TNF-alpha) stimulation. RNA polymerase II (Pol II) recruitment to the HIV-1 LTR closely mirrored RelA binding. Transient stimulation of cells with TNF-alpha for 15 min induced only a single round of RelA and RNA Pol II binding and failed to induce robust expression of latent HIV-1. Efficient formation of elongated HIV-1 transcripts required sustained induction by NF-kappaB, which promoted de novo synthesis of Tat. Cyclin-dependent kinase 9 (CDK9) and serine-2-phosphorylated RNA Pol II were rapidly recruited to the HIV-1 LTR after NF-kappaB induction; however, these elongating polymerase complexes were progressively dephosphorylated in the absence of Tat. Okadaic acid promoted sustained serine-2 phosphorylation of the C-terminal domain of RNA Pol II and stimulated efficient transcriptional elongation and HIV-1 expression in the absence of Tat. These findings underscore important differences between NF-kappaB and Tat stimulation of RNA Pol II elongation. While NF-kappaB binding to the HIV-1 LTR induces serial waves of efficient RNA Pol II initiation, elongation is impaired by the action of an okadaic acid-sensitive phosphatase that dephosphorylates the C-terminal domain of RNA Pol II. Conversely, the action of this phosphatase is overcome in the presence of Tat, promoting very efficient RNA Pol II elongation.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , VIH-1/fisiología , FN-kappa B/biosíntesis , Latencia del Virus/fisiología , Línea Celular , Productos del Gen tat/biosíntesis , Productos del Gen tat/genética , Humanos , FN-kappa B/fisiología , Provirus/fisiología , ARN Polimerasa II/fisiología , Factor de Transcripción ReIA/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
9.
EMBO J ; 25(1): 139-49, 2006 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-16319923

RESUMEN

Cells latently infected with HIV represent a currently insurmountable barrier to viral eradication in infected patients. Using the J-Lat human T-cell model of HIV latency, we have investigated the role of host factor binding to the kappaB enhancer elements of the HIV long terminal repeat (LTR) in the maintenance of viral latency. We show that NF-kappaB p50-HDAC1 complexes constitutively bind the latent HIV LTR and induce histone deacetylation and repressive changes in chromatin structure of the HIV LTR, changes that impair recruitment of RNA polymerase II and transcriptional initiation. Knockdown of p50 expression with specific small hairpin RNAs reduces HDAC1 binding to the latent HIV LTR and induces RNA polymerase II recruitment. Similarly, inhibition of histone deacetylase (HDAC) activity with trichostatin A promotes binding of RNA polymerase II to the latent HIV LTR. This bound polymerase complex, however, remains non-processive, generating only short viral transcripts. Synthesis of full-length viral transcripts can be rescued under these conditions by expression of Tat. The combination of HDAC inhibitors and Tat merits consideration as a new strategy for purging latent HIV proviruses from their cellular reservoirs.


Asunto(s)
Infecciones por VIH/genética , VIH-1/fisiología , Histona Desacetilasas/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Latencia del Virus/genética , Células Cultivadas , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Productos del Gen tat/metabolismo , Infecciones por VIH/inmunología , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Histona Desacetilasa 1 , Inhibidores de Histona Desacetilasas , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Subunidad p50 de NF-kappa B/genética , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Linfocitos T/virología , Transcripción Genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
10.
J Virol ; 80(21): 10407-18, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16956949

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) causes cell cycle arrest in G2. Vpr-expressing cells display the hallmarks of certain forms of DNA damage, specifically activation of the ataxia telangiectasia mutated and Rad3-related kinase, ATR. However, evidence that Vpr function is relevant in vivo or in the context of viral infection is still lacking. In the present study, we demonstrate that HIV-1 infection of primary, human CD4+ lymphocytes causes G2 arrest in a Vpr-dependent manner and that this response requires ATR, as shown by RNA interference. The event leading to ATR activation in CD4+ lymphocytes is the accumulation of replication protein A in nuclear foci, an indication that Vpr likely induces stalling of replication forks. Primary macrophages are refractory to ATR activation by Vpr, a finding that is consistent with the lack of detectable ATR, Rad17, and Chk1 protein expression in these nondividing cells. These observations begin to explain the remarkable resilience of macrophages to HIV-1-induced cytopathicity. To study the in vivo consequences of Vpr function, we isolated CD4+ lymphocytes from HIV-1-infected individuals and interrogated the cell cycle status of anti-p24Gag-immunoreactive cells. We report that infected cells in vivo display an aberrant cell cycle profile whereby a majority of cells have a 4N DNA content, consistent with the onset of G2 arrest.


Asunto(s)
Replicación del ADN , Productos del Gen vpr/fisiología , VIH-1/patogenicidad , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Línea Celular , Células Cultivadas , Efecto Citopatogénico Viral , ADN Viral/biosíntesis , ADN Viral/genética , Fase G2 , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Humanos , Técnicas In Vitro , Macrófagos/metabolismo , Macrófagos/virología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Interferencia de ARN , Transducción de Señal , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
11.
J Immunol ; 175(9): 6050-7, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16237100

RESUMEN

The HIV-1 protein Nef enhances viral pathogenicity and accelerates disease progression in vivo. Nef potentiates T cell activation by an unknown mechanism, probably by optimizing the intracellular environment for HIV replication. Using a new T cell reporter system, we have found that Nef more than doubles the number of cells expressing the transcription factors NF-kappaB and NFAT after TCR stimulation. This Nef-induced priming of TCR signaling pathways occurred independently of calcium signaling and involved a very proximal step before protein kinase C activation. Engagement of the TCR by MHC-bound Ag triggers the formation of the immunological synapse by recruiting detergent-resistant membrane microdomains, termed lipid rafts. Approximately 5-10% of the total cellular pool of Nef is localized within lipid rafts. Using confocal and real-time microscopy, we found that Nef in lipid rafts was recruited into the immunological synapse within minutes after Ab engagement of the TCR/CD3 and CD28 receptors. This recruitment was dependent on the N-terminal domain of Nef encompassing its myristoylation. Nef did not increase the number of cell surface lipid rafts or immunological synapses. Recently, studies have shown a specific interaction of Nef with an active subpopulation of p21-activated kinase-2 found only in the lipid rafts. Thus, the corecruitment of Nef and key cellular partners (e.g., activated p21-activated kinase-2) into the immunological synapse may underlie the increased frequency of cells expressing transcriptionally active forms of NF-kappaB and NFAT and the resultant changes in T cell activation.


Asunto(s)
Productos del Gen nef/fisiología , VIH/fisiología , Activación de Linfocitos , Microdominios de Membrana/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología , Antígenos CD28/fisiología , Señalización del Calcio , Productos del Gen nef/química , Humanos , Células Jurkat , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Estructura Terciaria de Proteína , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Quinasas p21 Activadas
12.
J Biol Chem ; 279(40): 42008-17, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15284245

RESUMEN

A subset of quiescent memory CD4 T cells harboring integrated but transcriptionally silent proviruses poses a currently insurmountable barrier to the eradication of the human immunodeficiency virus (HIV) in infected patients. Induction of HIV gene expression in these latently infected cells by immune activating agents has been proposed as one approach to confer sensitivity to antiretroviral therapy. Interest has recently focused on the non-tumor-promoting phorbol ester, prostratin, as a potential agent to activate latent HIV proviruses. Using multiple Jurkat T cell lines containing integrated but transcriptionally latent HIV proviruses (J-Lat cells), we now demonstrate that prostratin effectively activates HIV gene expression in these latently infected cells. We further show that prostratin acts by stimulating IKK-dependent phosphorylation and degradation of IkappaBalpha, leading to the rapid nuclear translocation of NF-kappaB and activation of the HIV-1 long terminal repeat in a kappaB enhancer-dependent manner. In contrast, NFAT and AP-1 are not induced by prostratin. Using chromatin immunoprecipitation assays to identify host transcription factors recruited to the latent HIV-1 promoter in living cells, we find that prostratin induces RelA binding. Analysis of potential upstream signal transducers demonstrates that prostratin stimulates membrane translocation of classical, novel, and atypical protein kinase C (PKC) isoforms. Studies with isoform-specific PKC inhibitors suggest that the novel PKCs play a particularly prominent role in the prostratin response. These findings provide new insights into the molecular pathway through which prostratin antagonizes HIV latency highlighting a central role for the action of NF-kappaB.


Asunto(s)
VIH/fisiología , FN-kappa B/metabolismo , Ésteres del Forbol/farmacología , Latencia del Virus/efectos de los fármacos , Transporte Activo de Núcleo Celular , Linfocitos T CD4-Positivos/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Duplicado del Terminal Largo de VIH , Humanos , Quinasa I-kappa B , Células Jurkat , FN-kappa B/efectos de los fármacos , Fosforilación , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo
13.
Virology ; 302(1): 95-105, 2002 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-12429519

RESUMEN

The 14-kDa Vpr protein of human immunodeficiency virus type 1 (HIV-1) serves multiple functions in the retroviral life cycle, including the enhancement of viral replication in nondividing macrophages, the induction of G2 cell-cycle arrest in proliferating T lymphocytes, and the modulation of HIV-1-induced apoptosis. Extracellular Vpr has been detected in the sera and cerebral spinal fluid of HIV-infected patients. However, it is not known whether such forms of Vpr are biologically active. Vpr contains a carboxy-terminal basic amino acid rich segment stretch that is homologous to domains that mediate the energy- and receptor-independent cellular uptake of polypeptides by a process termed protein transduction. Similar functional protein-transducing domains are present in HIV-1 Tat, herpes simplex virus-1 DNA-binding protein VP22, and the Drosophila antennapedia homeotic transcription factor. We now demonstrate effective transduction of biologically active, synthetic Vpr (sVpr) as well as the Vpr-beta-galactosidase fusion protein. However, in contrast to other transducing proteins, Vpr transduction is not enhanced by protein denaturation, and Vpr's carboxy-terminal basic domain alone is not sufficient for its transduction across biological membranes. In contrast, the full-length Vpr protein effectively transduces a broad array of cells, leading to dose-dependent G2 cell-cycle arrest and apoptosis. Addition of Vpr into the extracellular medium also rescues the replication of Vpr-deficient strains of HIV-1 in human macrophage cultures. Native Vpr may thus be optimized for protein transduction, a feature that might enhance and extend the pathological effects of HIV infection.


Asunto(s)
Productos del Gen vpr/metabolismo , VIH-1/metabolismo , Línea Celular Transformada , Productos del Gen vpr/genética , Células HeLa , Humanos , Células Jurkat , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
14.
J Virol ; 77(13): 7582-9, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12805458

RESUMEN

Retroviruses must gain access to the host cell nucleus for subsequent replication and viral propagation. Human immunodeficiency virus type 1 (HIV-1) and other primate lentiviruses are distinguished from the gammaretroviruses by their ability to infect nondividing cells such as macrophages, an important viral reservoir in vivo. Rather than requiring nuclear membrane breakdown during cell division, the HIV-1 preintegration complex (PIC) enters the nucleus by traversing the central aqueous channel of the limiting nuclear pore complex. The HIV-1 PIC contains three nucleophilic proteins, matrix, integrase, and Vpr, all of which have been implicated in nuclear targeting. The mechanism by which Vpr can display such nucleophilic properties and yet also be available for incorporation into virions assembling at the plasma membrane is unresolved. We recently characterized Vpr as a nucleocytoplasmic shuttling protein that contains two novel nuclear import signals and an exportin-1-dependent nuclear export signal (NES). We now demonstrate that mutation of this NES impairs the incorporation of Vpr into newly formed virions. Furthermore, we find that the Vpr NES is required for efficient HIV replication in tissue macrophages present in human spleens and tonsils. These findings underscore how the nucleocytoplasmic shuttling of Vpr not only contributes to nuclear import of the HIV-1 PIC but also enables Vpr to be present in the cytoplasm for incorporation into virions, leading to enhancement of viral spread within nondividing tissue macrophages.


Asunto(s)
Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Productos del Gen vpr/metabolismo , VIH-1/fisiología , Macrófagos/virología , Replicación Viral , Línea Celular , Fase G2 , Humanos , Tejido Linfoide/citología , Tejido Linfoide/virología , Transporte de Proteínas , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA