Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Retrovirology ; 16(1): 25, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492165

RESUMEN

Of the members of the primate T cell lymphotropic virus (PTLV) family, only the human T-cell leukemia virus type-1 (HTLV-1) causes disease in humans-as the etiological agent of adult T-cell leukemia/lymphoma (ATLL), HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and other auto-inflammatory disorders. Despite having significant genomic organizational and structural similarities, the closely related human T-cell lymphotropic virus type-2 (HTLV-2) is considered apathogenic and has been linked with benign lymphoproliferation and mild neurological symptoms in certain infected patients. The silencing of proviral gene expression and maintenance of latency are central for the establishment of persistent infections in vivo. The conserved pX sequences of HTLV-1 and HTLV-2 encode several ancillary factors which have been shown to negatively regulate proviral gene expression, while simultaneously activating host cellular proliferative and pro-survival pathways. In particular, the ORF-II proteins, HTLV-1 p30II and HTLV-2 p28II, suppress Tax-dependent transactivation from the viral promoter-whereas p30II also inhibits PU.1-mediated inflammatory-signaling, differentially augments the expression of p53-regulated metabolic/pro-survival genes, and induces lymphoproliferation which could promote mitotic proviral replication. The ubiquitinated form of the HTLV-1 p13II protein localizes to nuclear speckles and interferes with recruitment of the p300 coactivator by the viral transactivator Tax. Further, the antisense-encoded HTLV-1 HBZ and HTLV-2 APH-2 proteins and mRNAs negatively regulate Tax-dependent proviral gene expression and activate inflammatory signaling associated with enhanced T-cell lymphoproliferation. This review will summarize our current understanding of the pX latency-maintenance factors of HTLV-1 and HTLV-2 and discuss how these products may contribute to the differences in pathogenicity between the human PTLVs.


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 2 Humano/genética , Factores de Transcripción/genética , Proteínas Reguladoras y Accesorias Virales/genética , Latencia del Virus , Regulación Viral de la Expresión Génica , Infecciones por HTLV-I/complicaciones , Infecciones por HTLV-II/virología , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Virus Linfotrópico T Tipo 2 Humano/patogenicidad , Humanos , Virus Linfotrópico T Tipo 1 de los Primates/genética , Virus Linfotrópico T Tipo 1 de los Primates/patogenicidad , Proteínas Oncogénicas de Retroviridae/genética , Proteínas Oncogénicas de Retroviridae/metabolismo
2.
Virology ; 585: 1-20, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37257253

RESUMEN

The high-risk subtype human papillomaviruses (hrHPVs) infect and oncogenically transform basal epidermal stem cells associated with the development of squamous-cell epithelial cancers. The viral E6 oncoprotein destabilizes the p53 tumor suppressor, inhibits p53 K120-acetylation by the Tat-interacting protein of 60 kDa (TIP60, or Kat5), and prevents p53-dependent apoptosis. Intriguingly, the p53 gene is infrequently mutated in HPV + cervical cancer clinical isolates which suggests a possible paradoxical role for this gatekeeper in viral carcinogenesis. Here, we demonstrate that E6 activates the TP53-induced glycolysis and apoptosis regulator (TIGAR) and protects cells against oncogene-induced oxidative genotoxicity. The E6 oncoprotein induces a Warburg-like stress response and activates PI3K/PI5P4K/AKT-signaling that phosphorylates the TIGAR on serine residues and induces its hypoxia-independent mitochondrial targeting in hrHPV-transformed cells. Primary HPV + cervical cancer tissues contain high levels of TIGAR, p53, and c-Myc and our xenograft studies have further shown that lentiviral-siRNA-knockdown of TIGAR expression inhibits hrHPV-induced tumorigenesis in vivo. These findings suggest the modulation of p53 pro-survival signals and the antioxidant functions of TIGAR could have key ancillary roles during HPV carcinogenesis.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Femenino , Humanos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Virus del Papiloma Humano , Genes p53 , Neoplasias del Cuello Uterino/genética , Infecciones por Papillomavirus/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Glucólisis , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Carcinogénesis/genética , Hipoxia
3.
Artículo en Inglés | MEDLINE | ID: mdl-35291688

RESUMEN

The high-risk subtype Human Papillomaviruses (hrHPVs), including HPV16, HPV18, HPV31, HPV33, and HPV45, infect and oncogenically transform epithelial cells and cause squamous cell carcinomas and adenocarcinomas associated with the development of cervical cancer and subsets of vulvar, vaginal, penile, and anogenital cancers, as well as head-and-neck oropharyngeal carcinomas which often have poor clinical prognoses. Many cancers have been shown to contain elevated levels of the TP53-Induced Glycolysis and Apoptosis Regulator (TIGAR)-a glycolytic enzyme and antioxidant effector which frequently correlates with an aggressive tumor phenotype and serves as a determinant of therapy-resistance. We therefore tested whether siRNA-inhibition of TIGAR protein expression could sensitize HPV18-transformed HeLa cells to genotoxic chemotherapy agents (i.e., cisplatin, etoposide, doxorubicin, and 4-hydroxycyclophosphamide) that induce oxidative stress and DNA-damage. Here we demonstrate that the siRNA-knockdown of TIGAR hypersensitized HeLa cells to low, otherwise sub-inhibitory concentrations of these drugs and markedly induced cellular apoptosis, as compared to a scrambled RNA (scrRNA) oligonucleotide negative control or a non-transformed immortalized human fibroblast cell-line, HFL1. Importantly, these findings suggest that therapeutically inhibiting TIGAR could hypersensitize hrHPV+ cervical tumor cells to low-dosage concentrations of chemotherapy drugs that induce oxidative DNA-damage, which could potentially lead to more favorable clinical outcomes by reducing the adverse side-effects of these anticancer medications and making them more tolerable for patients. Our studies have further shown that siRNA-inhibition of TIGAR sensitizes HPV18+ HeLa cells to apoptosis induced by 4-hydroxycyclophosphamide-a DNA-alkylating agent these cells were reported to have resistance to, alluding to another possible benefit of targeting TIGAR in combinatorial treatment strategies against virus-induced cancers.

4.
J Exp Pharmacol ; 12: 503-515, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33262663

RESUMEN

Over the past 15 years, investigators have reported on the utility and safety of cardiac glycosides for numerous health benefits including those as treatments for malignant disease, stroke-mediated ischemic injury and certain neurodegenerative diseases. In addition to those, there is a growing body of evidence for novel antiviral effects of selected cardiac glycoside molecules. One unique cardiac glycoside, oleandrin derived from Nerium oleander, has been reported to have antiviral activity specifically against 'enveloped' viruses including HIV and HTLV-1. Importantly, a recent publication has presented in vitro evidence for oleandrin's ability to inhibit production of infectious virus particles when used for treatment prior to, as well as after infection by SARS-CoV-2/COVID-19. This review will highlight the known in vitro antiviral effects of oleandrin as well as present previously unpublished effects of this novel cardiac glycoside against Ebola virus, Cytomegalovirus, and Herpes simplex viruses.

5.
Artículo en Inglés | MEDLINE | ID: mdl-31824586

RESUMEN

At present, there are no antiretroviral drugs that inhibit incorporation of the envelope glycoprotein into newly-synthesized virus particles. The botanical glycoside, oleandrin, derived from extracts of Nerium oleander, has previously been shown to reduce the levels of the gp120 envelope glycoprotein on human immunodeficiency virus type-1 (HIV-1) particles and inhibit HIV-1 infectivity in vitro. We therefore tested whether oleandrin or an extract from N. oleander could also inhibit the infectivity of the human T-cell leukemia virus type-1 (HTLV-1): A related enveloped retrovirus and emerging tropical infectious agent. The treatment of HTLV-1+ lymphoma T-cells with either oleandrin or a N. oleander extract did not significantly inhibit viral replication or the release of p19Gag-containing particles into the culture supernatants. However, the collected virus particles from treated cells exhibited reduced infectivity on primary human peripheral blood mononuclear cells (huPBMCs). Unlike HIV-1, extracellular HTLV-1 particles are poorly infectious and viral transmission typically occurs via direct intercellular interactions across a virological synapse. We therefore investigated whether oleandrin or a N. oleander extract could inhibit virus transmission from a GFP-expressing HTLV-1+ lymphoma T-cell-line to huPBMCs in co-culture assays. These results demonstrated that both oleandrin and the crude phytoextract inhibited the formation of virological synapses and the transmission of HTLV-1 in vitro. Importantly, these findings suggest oleandrin may have broad antiviral activity against enveloped viruses by reducing the incorporation of the envelope glycoprotein into mature particles, a stage of the infection cycle not targeted by modern HAART.

6.
Virology ; 535: 83-101, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31299491

RESUMEN

Genomic instability is a hallmark of many cancers; however, the molecular etiology of chromosomal dysregulation is not well understood. The human T-cell leukemia virus type-1 (HTLV-1) oncoprotein Tax activates NF-κB-signaling and induces DNA-damage and aberrant chromosomal segregation through diverse mechanisms which contribute to viral carcinogenesis. Intriguingly, Stathmin/oncoprotein-18 (Op-18) depolymerizes tubulin and interacts with the p65RelA subunit and functions as a cofactor for NF-κB-dependent transactivation. We thus hypothesized that the dissociation of p65RelA-Stathmin/Op-18 complexes by Tax could lead to the catastrophic destabilization of microtubule (MT) spindle fibers during mitosis and provide a novel mechanistic link between NF-κB-signaling and genomic instability. Here we report that the inhibition of Stathmin expression by the retroviral latency protein, p30II, or knockdown with siRNA-stathmin, dampens Tax-mediated NF-κB transactivation and counters Tax-induced genomic instability and cytotoxicity. The Tax-G148V mutant, defective for NF-κB activation, exhibited reduced p65RelA-Stathmin binding and diminished genomic instability and cytotoxicity. Dominant-negative inhibitors of NF-κB also prevented Tax-induced multinucleation and apoptosis. Moreover, cell clones containing the infectious HTLV-1 ACH. p30II mutant provirus, impaired for p30II production, exhibited increased multinucleation and the accumulation of cytoplasmic tubulin aggregates following nocodozole-treatment. These findings allude to a mechanism whereby NF-κB-signaling regulates tubulin dynamics and mitotic instability through the modulation of p65RelA-Stathmin/Op-18 interactions, and support the notion that p30II enhances the survival of Tax-expressing HTLV-1-transformed cells.


Asunto(s)
Productos del Gen tax/metabolismo , Inestabilidad Genómica , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Huso Acromático/metabolismo , Estatmina/metabolismo , Factor de Transcripción ReIA/metabolismo , Células HEK293 , Humanos , Unión Proteica , Mapas de Interacción de Proteínas
7.
Mol Cell Biol ; 25(14): 6178-98, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15988028

RESUMEN

The human T-cell lymphotropic virus type 1 (HTLV-1) infects and transforms CD4+ lymphocytes and causes adult T-cell leukemia/lymphoma (ATLL), an aggressive lymphoproliferative disease that is often fatal. Here, we demonstrate that the HTLV-1 pX splice-variant p30II markedly enhances the transforming potential of Myc and transcriptionally activates the human cyclin D2 promoter, dependent upon its conserved Myc-responsive E-box enhancer elements, which are associated with increased S-phase entry and multinucleation. Enhancement of c-Myc transforming activity by HTLV-1 p30II is dependent upon the transcriptional coactivators, transforming transcriptional activator protein/p434 and TIP60, and it requires TIP60 histone acetyltransferase (HAT) activity and correlates with the stabilization of HTLV-1 p30II/Myc-TIP60 chromatin-remodeling complexes. The p30II oncoprotein colocalizes and coimmunoprecipitates with Myc-TIP60 complexes in cultured HTLV-1-infected ATLL patient lymphocytes. Amino acid residues 99 to 154 within HTLV-1 p30II interact with the TIP60 HAT, and p30II transcriptionally activates numerous cellular genes in a TIP60-dependent or TIP60-independent manner, as determined by microarray gene expression analyses. Importantly, these results suggest that p30II functions as a novel retroviral modulator of Myc-TIP60-transforming interactions that may contribute to adult T-cell leukemogenesis.


Asunto(s)
Acetiltransferasas/metabolismo , Ciclinas/genética , Elementos E-Box/genética , Leucemia-Linfoma de Células T del Adulto/virología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas de los Retroviridae/metabolismo , Activación Transcripcional , Acetiltransferasas/análisis , Empalme Alternativo , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/virología , Transformación Celular Neoplásica , Ensamble y Desensamble de Cromatina , Ciclina D2 , Perfilación de la Expresión Génica , Histona Acetiltransferasas , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Lisina Acetiltransferasa 5 , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteínas de los Retroviridae/análisis , Proteínas de los Retroviridae/genética , Transcripción Genética
8.
Virology ; 518: 103-115, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29462755

RESUMEN

In normal cells, aberrant oncogene expression leads to the accumulation of cytotoxic metabolites, including reactive oxygen species (ROS), which can cause oxidative DNA-damage and apoptosis as an intrinsic barrier against neoplastic disease. The c-Myc oncoprotein is overexpressed in many lymphoid cancers due to c-myc gene amplification and/or 8q24 chromosomal translocations. Intriguingly, p53 is a downstream target of c-Myc and hematological malignancies, such as adult T-cell leukemia/lymphoma (ATL), frequently contain wildtype p53 and c-Myc overexpression. We therefore hypothesized that p53-regulated pro-survival signals may thwart the cell's metabolic anticancer defenses to support oncogene-activation in lymphoid cancers. Here we show that the Tp53-induced glycolysis and apoptosis regulator (TIGAR) promotes c-myc oncogene-activation by the human T-cell leukemia virus type-1 (HTLV-1) latency-maintenance factor p30II, associated with c-Myc deregulation in ATL clinical isolates. TIGAR prevents the intracellular accumulation of c-Myc-induced ROS and inhibits oncogene-induced cellular senescence in ATL, acute lymphoblastic leukemia, and multiple myeloma cells with elevated c-Myc expression. Our results allude to a pivotal role for p53-regulated antioxidant signals as mediators of c-Myc oncogenic functions in viral and non-viral lymphoid tumors.


Asunto(s)
Carcinogénesis , Regulación Viral de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oncogenes/fisiología , Estrés Oxidativo/fisiología , Proteínas de los Retroviridae/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Reguladoras de la Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Monoéster Fosfórico Hidrolasas , Especies Reactivas de Oxígeno , Proteínas de los Retroviridae/genética
9.
Virology ; 520: 39-58, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29777913

RESUMEN

The human T-cell leukemia virus type-1 (HTLV-1) is an oncoretrovirus that infects and transforms CD4+ T-cells and causes adult T-cell leukemia/lymphoma (ATLL) -an aggressive lymphoproliferative disease that is highly refractive to most anticancer therapies. The HTLV-1 proviral genome encodes several regulatory products within a conserved 3' nucleotide sequence, known as pX; however, it remains unclear how these factors might cooperate or dynamically interact in virus-infected cells. Here we demonstrate that the HTLV-1 latency-maintenance factor p30II induces the TP53-induced glycolysis and apoptosis regulator (TIGAR) and counters the oxidative stress, mitochondrial damage, and cytotoxicity caused by the viral oncoproteins Tax and HBZ. The p30II protein cooperates with Tax and HBZ and enhances their oncogenic potential in colony transformation/foci-formation assays. Further, we have shown that TIGAR is highly expressed in HTLV-1-induced tumors associated with oncogene dysregulation and increased angiogenesis in an in vivo xenograft model of HTLV-1-induced T-cell lymphoma. These findings provide the first evidence that p30II likely collaborates as an ancillary factor for the major oncoproteins Tax and HBZ during retroviral carcinogenesis.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Linfoma/virología , Proteínas de los Retroviridae/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Carcinogénesis , Regulación Viral de la Expresión Génica , Genes pX , Xenoinjertos , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Mitofagia , Neovascularización Patológica , Estrés Oxidativo , Monoéster Fosfórico Hidrolasas , Especies Reactivas de Oxígeno/metabolismo , Proteínas de los Retroviridae/genética
10.
Oncogene ; 24(39): 6026-34, 2005 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16155609

RESUMEN

The human T-cell leukemia/lymphoma virus (HTLV) genome, in addition to the structural Gag and Env proteins and retroviral enzymes, carries a region at its 3' end originally designated pX. To date, we know that this region encodes two essential transcriptional and post-transcriptional positive regulators of viral expression, the Tax and Rex proteins, respectively (reviewed elsewhere in this issue). Here, we will review current knowledge of the functions of three additional proteins encoded in the pX region, p12I, p13II, and p30II.


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano/genética , Proteínas no Estructurales Virales/genética , Secuencia de Aminoácidos , Genoma Viral , Modelos Biológicos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Procesamiento Postranscripcional del ARN , Transcripción Genética
11.
Virology ; 476: 271-288, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25569455

RESUMEN

The human T-cell leukemia retrovirus type-1 (HTLV-1) p30(II) protein is a multifunctional latency-maintenance factor that negatively regulates viral gene expression and deregulates host signaling pathways involved in aberrant T-cell growth and proliferation. We have previously demonstrated that p30(II) interacts with the c-MYC oncoprotein and enhances c-MYC-dependent transcriptional and oncogenic functions. However, the molecular and biochemical events that mediate the cooperation between p30(II) and c-MYC remain to be completely understood. Herein we demonstrate that p30(II) induces lysine-acetylation of the c-MYC oncoprotein. Acetylation-defective c-MYC Lys→Arg substitution mutants are impaired for oncogenic transformation with p30(II) in c-myc(-/-) HO15.19 fibroblasts. Using dual-chromatin-immunoprecipitations (dual-ChIPs), we further demonstrate that p30(II) is present in c-MYC-containing nucleoprotein complexes in HTLV-1-transformed HuT-102 T-lymphocytes. Moreover, p30(II) inhibits apoptosis in proliferating cells expressing c-MYC under conditions of genotoxic stress. These findings suggest that c-MYC-acetylation is required for the cooperation between p30(II)/c-MYC which could promote proviral replication and contribute to HTLV-1-induced carcinogenesis.


Asunto(s)
Transformación Celular Neoplásica , Infecciones por HTLV-I/metabolismo , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas de los Retroviridae/metabolismo , Acetilación , Secuencias de Aminoácidos , Proliferación Celular , Transformación Celular Viral , Infecciones por HTLV-I/genética , Infecciones por HTLV-I/fisiopatología , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas de los Retroviridae/genética
12.
J Biol Chem ; 282(29): 20854-67, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17526487

RESUMEN

Bovine leukemia virus (BLV) expression is controlled at the transcriptional level through three Tax(BLV)-responsive elements (TxREs) responsive to the viral transactivator Tax(BLV). The cAMP-responsive element (CRE)-binding protein (CREB) has been shown to interact with CRE-like sequences present in the middle of each of these TxREs and to play critical transcriptional roles in both basal and Tax(BLV)-transactivated BLV promoter activity. In this study, we have investigated the potential involvement of the cAMP-response element modulator (CREM) in BLV transcriptional regulation, and we have demonstrated that CREM proteins were expressed in BLV-infected cells and bound to the three BLV TxREs in vitro. Chromatin immunoprecipitation assays using BLV-infected cell lines demonstrated in the context of chromatin that CREM proteins were recruited to the BLV promoter TxRE region in vivo. Functional studies, in the absence of Tax(BLV), indicated that ectopic CREMtau protein had a CRE-dependent stimulatory effect on BLV promoter transcriptional activity. Cross-link of the B-cell receptor potentiated CREMtau transactivation of the viral promoter. Further experiments supported the notion that this potentiation involved CREMtau Ser-117 phosphorylation and recruitment of CBP/p300 to the BLV promoter. Although CREB and Tax(BLV) synergistically transactivated the BLV promoter, CREMtau repressed this Tax(BLV)/CREB synergism, suggesting that a modulation of the level of Tax(BLV) transactivation through opposite actions of CREB and CREMtau could facilitate immune escape and allow tumor development.


Asunto(s)
Modulador del Elemento de Respuesta al AMP Cíclico/genética , Virus de la Leucemia Bovina/genética , Transcripción Genética , Animales , Núcleo Celular/metabolismo , Clonación Molecular , Reactivos de Enlaces Cruzados/farmacología , AMP Cíclico/metabolismo , Modulador del Elemento de Respuesta al AMP Cíclico/química , Productos del Gen tax/metabolismo , Virus de la Leucemia Bovina/metabolismo , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Fosforilación , Regiones Promotoras Genéticas , Isoformas de Proteínas
13.
J Biol Chem ; 282(16): 12048-57, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17317667

RESUMEN

The Werner syndrome helicase (WRN) participates in DNA replication, double strand break repair, telomere maintenance, and p53 activation. Mutations of wrn cause Werner syndrome (WS), an autosomal recessive premature aging disorder associated with cancer predisposition, atherosclerosis, and other aging related symptoms. Here, we report that WRN is a novel cofactor for HIV-1 replication. Immortalized human WRN(-/-) WS fibroblasts, lacking a functional wrn gene, are impaired for basal and Tat-activated HIV-1 transcription. Overexpression of wild-type WRN transactivates the HIV-1 long terminal repeat (LTR) in the absence of Tat, and WRN cooperates with Tat to promote high-level LTR transactivation. Ectopic WRN induces HIV-1 p24(Gag) production and retroviral replication in HIV-1-infected H9(HIV-1IIIB) lymphocytes. A dominant-negative helicase-minus mutant, WRN(K577M), inhibits LTR transactivation and HIV-1 replication. Inhibition of endogenous WRN, through co-expression of WRN(K577M), diminishes recruitment of p300/CREB-binding protein-associated factor (PCAF) and positive transcription elongation factor b (P-TEFb) to Tat/transactivation response-RNA complexes, and immortalized WRN(-/-) WS fibroblasts exhibit comparable defects in recruitment of PCAF and P-TEFb to the HIV-1 LTR. Our results demonstrate that WRN is a novel cellular cofactor for HIV-1 replication and suggest that the WRN helicase participates in the recruitment of PCAF/P-TEFb-containing transcription complexes. WRN may be a plausible target for antiretroviral therapy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Helicasas/metabolismo , Duplicado del Terminal Largo de VIH , Histona Acetiltransferasas/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , RecQ Helicasas/fisiología , Retroviridae/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Replicación Viral , Exodesoxirribonucleasas , Fibroblastos/metabolismo , Productos del Gen tat/metabolismo , Células HeLa , Humanos , Linfocitos/metabolismo , Modelos Biológicos , RecQ Helicasas/metabolismo , Síndrome de Werner/enzimología , Helicasa del Síndrome de Werner , Factores de Transcripción p300-CBP
15.
J Biol Chem ; 280(10): 9390-9, 2005 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-15611041

RESUMEN

The human immunodeficiency virus type-1 (HIV-1) infects microglia, macrophages, and astrocytes in the central nervous system (CNS) and may cause severe neurological diseases, such as AIDS-related dementias or progressive encephalopathies, as a result of CNS inflammation and neurotrophin signaling defects associated with expression of viral antigens and HIV-1 replication in the brain. The HIV Tat protein can be endocytosed by surrounding uninfected cells; interacts with transcriptional coactivators/acetyltransferases, p300/CREB-binding protein, and p300/CREB-binding protein-associated factor (PCAF); and induces neuronal apoptosis. Since nerve growth factor (NGF) receptor and brain-derived neurotrophic factor receptor signaling through CREB requires p300 and PCAF histone acetyltransferases, we sought to determine whether HIV-1 Tat coactivator interactions interfere with neurotrophin receptor signaling in neuronal cells. Here, we demonstrate that Tat-coactivator interactions inhibit NGF- and brain-derived neurotrophic factor-responsive CRE trans-activation and neurotrophin protection against apoptosis in PC12 and IMR-32 neuroblastoma cells. Purified recombinant Tat or Tat-derived synthetic peptides, spanning p300- and PCAF-binding sequences, inhibit histone H3/H4 acetylation in vitro. A Tat mutant, TatK28A/K50A, defective for binding p300 and PCAF, neither repressed NGF-responsive CRE transactivation nor inhibited histone acetylation. HIV-1 Tat interacts in PCAF complexes in post-mortem CNS tissues from donor neuro-AIDS patients, as determined by fluorescence resonance energy transfer immunoconfocal microscopy. Importantly, these findings suggest that HIV-1 Tat-coactivator interactions may contribute to neurotrophin signaling impairments and neuronal apoptosis associated with HIV-1 infections of the CNS.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Productos del Gen tat/metabolismo , Histonas/metabolismo , Factores de Crecimiento Nervioso/fisiología , Factores de Transcripción/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Transferencia Resonante de Energía de Fluorescencia , VIH-1/fisiología , Histona Acetiltransferasas , Humanos , Microscopía Confocal , Datos de Secuencia Molecular , Neuroblastoma , Células PC12 , Feocromocitoma , Ratas , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección , Replicación Viral , Factores de Transcripción p300-CBP , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
16.
J Biol Chem ; 278(14): 12310-8, 2003 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-12501250

RESUMEN

Patients with AIDS are at increased risk for developing various neoplasms, including Hodgkin's and non-Hodgkin's lymphomas, Kaposi's sarcomas, and anal-rectal carcinomas, suggestive that human immunodeficiency virus type-1 infection might promote establishment of AIDS-related cancers. Tat, the viral trans-activator, can be endocytosed by uninfected cells and has been shown to inhibit p53 functions, providing a candidate mechanism through which the human immunodeficiency virus type-1 might contribute to malignant transformation. Because Tat has been shown to interact with histone acetyltransferase domains of p300/cAMP-responsive element-binding protein (CREB)-binding protein and p300/CREB-binding protein-associated factor, we have investigated whether Tat might alter p53 acetylation and tumor suppressor-responsive transcription. Here, we demonstrate that both Tat and p53 co-localize with p300/CREB-binding protein-associated factor and p300 in nuclei of IMR-32 human neuroblastoma cells and in PC-12 pheochromocytoma cells. Further, p53 trans-activation of the 14-3-3varsigma promoter was markedly repressed by Tat-histone acetyltransferase interactions, and p53 acetylation by p300/CREB-binding protein-associated factor on residue Lys(320) was diminished as a result of Tat-histone acetyltransferase binding in vivo and in vitro. Tat also inhibited p53 acetylation by p300 in a dosage-dependent manner in vitro. Finally, HIV-1-infected Molt-4 cells displayed reduced p53 acetylation on lysines 320 and 373 in response to UV irradiation. Our results allude to a mechanism whereby the human immunodeficiency virus type-1 trans-activator might impair tumor suppressor functions in immune/neuronal-derived cells, thus favoring the establishment of neoplasia during AIDS.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Productos del Gen tat/metabolismo , VIH-1 , VIH/metabolismo , Activación Transcripcional/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de la radiación , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Núcleo Celular/metabolismo , Doxorrubicina/farmacología , Productos del Gen tat/genética , Histona Acetiltransferasas , Humanos , Neuroblastoma , Células PC12 , Fragmentos de Péptidos/metabolismo , Regiones Promotoras Genéticas/fisiología , Ratas , Transactivadores/metabolismo , Factores de Transcripción , Transfección , Células Tumorales Cultivadas , Rayos Ultravioleta , Factores de Transcripción p300-CBP , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
17.
Vaccine ; 20(25-26): 3171-86, 2002 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12163269

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) regulatory proteins Rev, Tat, and Nef are expressed at early time post-infection and represent attractive targets to be included in a vaccine candidate for AIDS. However, the putative immunosuppressive activities of some of these proteins may limit their immunogenicity. To circumvent these issues, a novel chimeric polyprotein vaccine candidate (Retanef), comprising genetically modified and re-assorted rev, tat, and nef open reading frames of simian immunodeficiency virus (SIV), was constructed and optimized for its expression in mammalian cells. Retanef encodes a protein of approximately 55 kDa localized primarily in the cytoplasm of transfected cells. The Retanef gene expressed in context of an eucaryotic expression vector (DNA-SIV-Retanef) or cloned into a highly attenuated poxvirus-based NYVAC vector (NYVAC-SIV-Retanef) was used to immunize either naive rhesus macaques or macaques chronically infected with SIVmac251 undergoing anti-retroviral therapy (ART). Three immunizations of naive macaques with DNA-SIV-Retanef followed by a single NYVAC-SIV-Retanef boost induced a response to the Mamu-A(*)01-restricted Tat epitope (Tat_SL8, TTPESANL) demonstrated by staining with a specific tetramer and by direct cytolytic activity assays, as well as responses to Rev, Tat and Nef proteins demonstrated by ELISPOT assays using overlapping peptide pools encompassing the entire proteins. Immunization of infected macaques with either DNA-SIV-Retanef or NYVAC-SIV-Retanef expanded the frequency of Tat-specific tetramer-staining cells by two- to seven-fold. No adverse effects were observed in either naive or SIV-infected rhesus macaques. Thus, an analogous HIV-1-based chimeric vaccine may represent useful component of an HIV-1 vaccine.


Asunto(s)
Vacunas contra el SIDA/genética , Genes nef , Genes rev , Genes tat , VIH-1/inmunología , Inmunotoxinas/genética , Vacunas contra el SIDAS/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas contra el SIDA/efectos adversos , Vacunas contra el SIDA/biosíntesis , Vacunas contra el SIDA/inmunología , Secuencia de Aminoácidos , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Secuencia de Bases , Chlorocebus aethiops , Genes env , Vectores Genéticos/genética , Antígenos VIH/genética , Antígenos VIH/inmunología , VIH-1/genética , Células HeLa , Humanos , Inmunotoxinas/inmunología , Macaca mulatta , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra el SIDAS/efectos adversos , Vacunas contra el SIDAS/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Transfección , Vacunación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Células Vero
18.
J Biol Chem ; 279(53): 55667-74, 2004 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-15496412

RESUMEN

The transcriptional coactivators, p300/CREB-binding protein-associated factor (PCAF) and hGCN5, are recruited to chromatin-remodeling complexes on enhancers of various gene promoters in response to growth factor stimulation. However, the molecular mechanisms by which surface receptor signals modulate the assembly of nuclear transcription complexes are not fully understood. Here we report that nerve growth factor receptor signaling induces nuclear translocation of PCAF and hGCN5 dependent upon the phosphorylation of Ser and Thr residues within their histone acetyltransferase domains, which requires activation of PI3K, Rsk2(pp90), and MSK-1. Neurotrophin stimulation induces p53(K320) acetylation by PCAF and transcriptionally activates p53-responsive enhancer elements within the p21(WAF/CIP1) promoter associated with G(1)/S arrest during neuronal differentiation. Most importantly, these findings represent the first evidence for signal-dependent nuclear translocation of PCAF and hGCN5 acetyltransferases and allude to a novel mechanism for ligand/receptor modulation of nuclear chromatin-remodeling complexes in neurons.


Asunto(s)
Acetiltransferasas/química , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transactivadores/metabolismo , Transporte Activo de Núcleo Celular , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Núcleo Celular/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Histona Acetiltransferasas , Humanos , Ligandos , Microscopía Confocal , Microscopía Fluorescente , Modelos Biológicos , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Células PC12 , Fosforilación , Estructura Terciaria de Proteína , Ratas , Transducción de Señal , Factores de Tiempo , Factores de Transcripción , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Factores de Transcripción p300-CBP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA