Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
2.
Cancer Cell ; 34(2): 286-297.e10, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30057145

RESUMEN

Adult T cell leukemia/lymphoma (ATLL) is a frequently incurable disease associated with the human lymphotropic virus type I (HTLV-I). RNAi screening of ATLL lines revealed that their proliferation depends on BATF3 and IRF4, which cooperatively drive ATLL-specific gene expression. HBZ, the only HTLV-I encoded transcription factor that is expressed in all ATLL cases, binds to an ATLL-specific BATF3 super-enhancer and thereby regulates the expression of BATF3 and its downstream targets, including MYC. Inhibitors of bromodomain-and-extra-terminal-domain (BET) chromatin proteins collapsed the transcriptional network directed by HBZ and BATF3, and were consequently toxic for ATLL cell lines, patient samples, and xenografts. Our study demonstrates that the HTLV-I oncogenic retrovirus exploits a regulatory module that can be attacked therapeutically with BET inhibitors.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Redes Reguladoras de Genes , Virus Linfotrópico T Tipo 1 Humano/fisiología , Factores Reguladores del Interferón/genética , Leucemia-Linfoma de Células T del Adulto/genética , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Línea Celular Tumoral , Genes myc , Humanos , Ratones , Proteínas/antagonistas & inhibidores , Proteínas de los Retroviridae/fisiología
3.
Leukemia ; 31(10): 2235-2243, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28260789

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) basic-leucine zipper (bZIP) factor (HBZ) is a key player in proliferation and transformation of HTLV-1-infected cells, thus contributing to adult T-cell leukemia (ATL) development. HBZ deregulates gene expression within the host cell by interacting with several cellular partners. Through its C-terminal ZIP domain, HBZ is able to contact and activate JunD, a transcription factor of the AP-1 family. JunD mRNA is intronless but can generate two protein isoforms by alternative translation initiation: JunD full-length and Δ JunD, an N-terminal truncated form unresponsive to the tumor suppressor menin. Using various cell lines and primary T-lymphocytes, we show that after serum deprivation HBZ induces the expression of Δ JunD isoform. We demonstrate that, unlike JunD, Δ JunD induces proliferation and transformation of cells. To decipher the mechanisms for Δ JunD production, we looked into the translational machinery and observed that HBZ induces nuclear retention of RPS25 mRNA and loss of RPS25 protein expression, a component of the small ribosomal subunit. Therefore, HBZ bypasses translational control of JunD uORF and favors the expression of Δ JunD. In conclusion, we provide strong evidences that HBZ induces Δ JunD expression through alteration of the cellular translational machinery and that the truncated isoform Δ JunD has a central role in the oncogenic process leading to ATL.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Transformación Celular Viral/genética , Regulación Leucémica de la Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Biosíntesis de Proteínas/genética , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas de los Retroviridae/fisiología , Proteínas Ribosómicas/antagonistas & inhibidores , Transporte Biológico , Línea Celular , Núcleo Celular/metabolismo , Medio de Cultivo Libre de Suero , Células HEK293 , Infecciones por HTLV-I/sangre , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-jun/genética , ARN Mensajero/metabolismo , Proteínas Ribosómicas/genética , Ribosomas/metabolismo , Linfocitos T/patología , Linfocitos T/virología , Transfección
4.
J Clin Invest ; 126(6): 2295-307, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27159392

RESUMEN

Current antiretroviral therapy (ART) is not sufficient to completely suppress disease progression in the CNS, as indicated by the rising incidence of HIV-1-associated neurocognitive disorders (HAND) among infected individuals on ART. It is not clear why some HIV-1-infected patients develop HAND, despite effective repression of viral replication in the circulation. SIV-infected nonhuman primate models are widely used to dissect the mechanisms of viral pathogenesis in the CNS. Here, we identified 4 amino acid substitutions in the cytoplasmic tail of viral envelope glycoprotein gp41 of the neurovirulent virus SIVsm804E that enhance replication in macrophages and associate with enhanced antagonism of the host restriction factor BM stromal cell antigen 2 (BST-2). Rhesus macaques were inoculated with a variant of the parental virus SIVsmE543-3 that had been engineered to contain the 4 amino acid substitutions present in gp41 of SIVsm804E. Compared with WT virus-infected controls, animals infected with mutant virus exhibited higher viral load in cerebrospinal fluid. Together, these results are consistent with a potential role for BST-2 in the CNS microenvironment and suggest that BST-2 antagonists may serve as a possible target for countermeasures against HAND.


Asunto(s)
Virus de la Inmunodeficiencia de los Simios/patogenicidad , Complejo SIDA Demencia/etiología , Sustitución de Aminoácidos , Animales , Antígenos CD/fisiología , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/fisiología , VIH-1 , Interacciones Huésped-Patógeno , Humanos , Macaca mulatta , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/fisiología , Carga Viral , Virulencia/genética , Replicación Viral/genética
5.
Oncogene ; 35(34): 4509-17, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-26804169

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) is an oncogenic retrovirus that induces a fatal T-cell malignancy, adult T-cell leukemia (ATL). Among several regulatory/accessory genes in HTLV-1, HTLV-1 bZIP factor (HBZ) is the only viral gene constitutively expressed in infected cells. Our previous study showed that HBZ functions in two different molecular forms, HBZ protein and HBZ RNA. In this study, we show that HBZ protein targets retinoblastoma protein (Rb), which is a critical tumor suppressor in many types of cancers. HBZ protein interacts with the Rb/E2F-1 complex and activates the transcription of E2F-target genes associated with cell cycle progression and apoptosis. Mouse primary CD4(+) T cells transduced with HBZ show accelerated G1/S transition and apoptosis, and importantly, T cells from HBZ transgenic (HBZ-Tg) mice also demonstrate enhanced cell proliferation and apoptosis. To evaluate the functions of HBZ protein alone in vivo, we generated a new transgenic mouse strain that expresses HBZ mRNA altered by silent mutations but encoding intact protein. In these mice, the numbers of effector/memory and Foxp3(+) T cells were increased, and genes associated with proliferation and apoptosis were upregulated. This study shows that HBZ protein promotes cell proliferation and apoptosis in primary CD4(+) T cells through activation of the Rb/E2F pathway, and that HBZ protein also confers onto CD4(+) T-cell immunophenotype similar to those of ATL cells, suggesting that HBZ protein has important roles in dysregulation of CD4(+) T cells infected with HTLV-1.


Asunto(s)
Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Linfocitos T CD4-Positivos/patología , Proteína de Retinoblastoma/fisiología , Proteínas de los Retroviridae/fisiología , Transducción de Señal/fisiología , Animales , Linfocitos T CD4-Positivos/virología , Ciclo Celular , Proliferación Celular , Células Cultivadas , Factor de Transcripción E2F1/fisiología , Histona Desacetilasas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/fisiología
6.
Cancer Res ; 75(19): 4143-52, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26383166

RESUMEN

Infection of T cells with human T-cell leukemia virus type-1 (HTLV-1) induces clonal proliferation and is closely associated with the onset of adult T-cell leukemia-lymphoma (ATL) and inflammatory diseases. Although Tax expression is frequently suppressed in HTLV-1-infected cells, the accessory gene, HTLV-1 bZIP factor (HBZ), is continuously expressed and has been implicated in HTLV-1 pathogenesis. Here, we report that transduction of mouse T cells with specific mutants of HBZ that distinguish between its RNA and protein activity results in differential effects on T-cell proliferation and survival. HBZ RNA increased cell number by attenuating apoptosis, whereas HBZ protein induced apoptosis. However, both HBZ RNA and protein promoted S-phase entry of T cells. We further identified that the first 50 bp of the HBZ coding sequence are required for RNA-mediated cell survival. Transcriptional profiling of T cells expressing wild-type HBZ, RNA, or protein revealed that HBZ RNA is associated with genes involved in cell cycle, proliferation, and survival, while HBZ protein is more closely related to immunological properties of T cells. Specifically, HBZ RNA enhances the promoter activity of survivin, an inhibitor of apoptosis, to upregulate its expression. Inhibition of survivin using YM155 resulted in impaired proliferation of several ATL cell lines as well as a T-cell line expressing HBZ RNA. The distinct functions of HBZ RNA and protein may have several implications for the development of strategies to control the proliferation and survival mechanisms associated with HTLV-1 infection and ATL.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Regulación Viral de la Expresión Génica , Virus Linfotrópico T Tipo 1 Humano/fisiología , ARN Viral/fisiología , Proteínas de los Retroviridae/fisiología , Linfocitos T/virología , Animales , Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , División Celular , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Imidazoles/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/fisiología , Leucemia-Linfoma de Células T del Adulto/patología , Ratones , Ratones Endogámicos C57BL , Mutación , Naftoquinonas/farmacología , Regiones Promotoras Genéticas , ARN/genética , ARN Viral/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Proteínas de los Retroviridae/genética , Survivin , Linfocitos T/fisiología , Transcripción Genética , Transducción Genética
7.
J Virol ; 89(10): 5450-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25741012

RESUMEN

UNLABELLED: Walleye dermal sarcoma virus (WDSV) infection is associated with the seasonal development and regression of walleye dermal sarcoma. Previous work showed that the retroviral cyclin (RV-cyclin), encoded by WDSV, has separable cyclin box and transcription activation domains. It binds to cyclin-dependent kinase 8 (CDK8) and enhances its kinase activity. CDK8 is evolutionarily conserved and is frequently overexpressed in human cancers. It is normally activated by cyclin C and is required for transcription elongation of the serum response genes (immediate early genes [IEGs]) FOS, EGR1, and cJUN. The IEGs drive cell proliferation, and their expression is brief and highly regulated. Here we show that constitutive expression of RV-cyclin in the HCT116 colon cancer cell line significantly increases the level of IEG expression in response to serum stimulation. Quantitative reverse transcription-PCR (RT-PCR) and nuclear run-on assays provide evidence that RV-cyclin does not alter the initiation of IEG transcription but does enhance the overall rate of transcription elongation and maintains transcription reinitiation. RV-cyclin does not increase activating phosphorylation events in the mitogen-activated protein kinase pathway and does not inhibit decay of IEG mRNAs. At the EGR1 gene locus, RV-cyclin increases and maintains RNA polymerase II (Pol II) occupancy after serum stimulation, in conjunction with increased and extended EGR1 gene expression. The RV-cyclin increases CDK8 occupancy at the EGR1 gene locus before and after serum stimulation. Both of RV-cyclin's functional domains, i.e., the cyclin box and the activation domain, are necessary for the overall enhancement of IEG expression. RV-cyclin presents a novel and ancient mechanism of retrovirus-induced oncogenesis. IMPORTANCE: The data reported here are important to both virology and cancer biology. The novel mechanism pinpoints CDK8 in the development of walleye dermal sarcoma and sheds light on CDK8's role in many human cancers. CDK8 controls expression from highly regulated genes, including the interferon-stimulated genes. Its function is likely the target of many viral interferon-resistance mechanisms. CDK8 also controls cellular responses to metabolic stimuli, stress, and hypoxia, in addition to the serum response. The retroviral cyclin (RV-cyclin) represents a highly selected probe of CDK8 function. RV-cyclin does not control CDK8 specificity but instead enhances CDK8's effects on regulated genes, an important distinction for its use to delineate natural CDK8 targets. The outcomes of this research are applicable to investigations of normal and abnormal CDK8 functions. The mechanisms defined here will contribute directly to the dermal sarcoma model in fish and clarify an important path for oncogenesis and innate resistance to viruses.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/metabolismo , Ciclinas/fisiología , Epsilonretrovirus/fisiología , Proteínas de los Retroviridae/fisiología , Animales , Carcinogénesis , Ciclinas/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Epsilonretrovirus/genética , Epsilonretrovirus/patogenicidad , Enfermedades de los Peces/genética , Enfermedades de los Peces/virología , Genes Inmediatos-Precoces , Genes fos , Genes jun , Células HCT116 , Interacciones Huésped-Patógeno , Humanos , Percas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/veterinaria , Infecciones por Retroviridae/virología , Proteínas de los Retroviridae/genética , Elongación de la Transcripción Genética , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/veterinaria , Infecciones Tumorales por Virus/virología
8.
Trends Microbiol ; 23(6): 367-75, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25701112

RESUMEN

Over recent years, retroviral gene expression has been shown to depend on a promoter that is bidirectional. This promoter activity is likely to occur at either end of the retroviral genome and has important consequences at the level of retroviral gene expression. This review focuses on the recent discovery of retroviral antisense genes termed HBZ [in human T-cell leukemia virus type 1 (HTLV-1)] and ASP (in HIV-1) in terms of their function and the regulation of their expression, both of which are interconnected with the expression and function of other viral proteins. Emphasis is also given to the potential implication of these proteins in the maintenance of chronic infection in infected individuals. In light of recent findings, the discovery of these new genes opens a new avenue for the future treatment of HTLV-1- and HIV-1-infected individuals.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Infecciones por VIH/virología , VIH-1/genética , Infecciones por HTLV-I/virología , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Virus Linfotrópico T Tipo 1 Humano/genética , Proteínas de los Retroviridae/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Enfermedad Crónica , VIH-1/fisiología , Proteínas del Virus de la Inmunodeficiencia Humana/fisiología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Regiones Promotoras Genéticas , Proteínas de los Retroviridae/fisiología , Transcripción Genética
9.
Bull Soc Pathol Exot ; 104(3): 181-7, 2011 Aug.
Artículo en Francés | MEDLINE | ID: mdl-21607661

RESUMEN

HTLV-1 was the first human oncogenic retrovirus to be discovered. It is the etiological agent of adult T leukemia/lymphoma (ATLL) and of tropical spastic paraparesis/HTLV-1 associated myelopathy (TSP/HAM), two diseases that develop after a long latency period. Importantly, HTLV-1 does not cause ATLL through insertional mutagenesis. Apart from the gag, pro, pol and env genes, which are common to all retroviruses, HTLV-1 genome also encodes regulatory and auxiliary viral proteins. Among the former, Tax promotes cell transformation and HBZ is involved in the leukemic cells proliferation and in the maintenance of the transformed phenotype. Anti-ATLL therapies have lately made significant progress with an efficient antiviral treatment against the chronic and smoldering forms of this leukemia, but an efficient treatment of TSP/HAM patients is still lacking. Results from a recent study associating histone acetylase inhibitor with an anti-viral drug will be discussed here. While an increase in proviral load is considered a marker for disease progression, this treatment allows a significant drop of the proviral load in asymptomatic carriers.


Asunto(s)
Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Animales , Antirretrovirales/uso terapéutico , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Genoma Viral , Infecciones por HTLV-I/tratamiento farmacológico , Infecciones por HTLV-I/prevención & control , Inhibidores de Histona Desacetilasas/uso terapéutico , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/virología , Paraparesia Espástica Tropical/tratamiento farmacológico , Paraparesia Espástica Tropical/virología , Primates , Provirus/efectos de los fármacos , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Ácido Valproico/uso terapéutico , Carga Viral , Replicación Viral
10.
PLoS Pathog ; 7(2): e1001268, 2011 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-21304939

RESUMEN

Refolding of viral class-1 membrane fusion proteins from a native state to a trimer-of-hairpins structure promotes entry of viruses into cells. Here we present the structure of the bovine leukaemia virus transmembrane glycoprotein (TM) and identify a group of asparagine residues at the membrane-distal end of the trimer-of-hairpins that is strikingly conserved among divergent viruses. These asparagines are not essential for surface display of pre-fusogenic envelope. Instead, substitution of these residues dramatically disrupts membrane fusion. Our data indicate that, through electrostatic interactions with a chloride ion, the asparagine residues promote assembly and profoundly stabilize the fusion-active structures that are required for viral envelope-mediated membrane fusion. Moreover, the BLV TM structure also reveals a charge-surrounded hydrophobic pocket on the central coiled coil and interactions with basic residues that cluster around this pocket are critical to membrane fusion and form a target for peptide inhibitors of envelope function. Charge-surrounded pockets and electrostatic interactions with small ions are common among class-1 fusion proteins, suggesting that small molecules that specifically target such motifs should prevent assembly of the trimer-of-hairpins and be of value as therapeutic inhibitors of viral entry.


Asunto(s)
Iones/metabolismo , Pliegue de Proteína , Proteínas de los Retroviridae/química , Proteínas de los Retroviridae/fisiología , Electricidad Estática , Secuencia de Aminoácidos , Animales , Antirretrovirales/química , Antirretrovirales/farmacología , Dominio Catalítico/efectos de los fármacos , Bovinos , Virus Linfotrópico T Tipo 1 Humano/química , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Enlace de Hidrógeno , Iones/química , Virus de la Leucemia Bovina/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/fisiología , Retroviridae/metabolismo , Retroviridae/fisiología , Proteínas de los Retroviridae/metabolismo , Propiedades de Superficie , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
11.
Blood ; 116(19): 3809-17, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20647569

RESUMEN

The identification of the genes necessary for human T-cell leukemia virus (HTLV-1) persistence in humans may provide targets for therapeutic approaches. We demonstrate that ablation of the HTLV-1 genes encoding p12, p30, or the HBZ protein, does not affect viral infectivity in rabbits and in this species, only the absence of HBZ is associated with a consistent reduction in virus levels. We observed reversion of the HTLV-1 mutants to the HTLV-1 wild-type genotype in none of the inoculated rabbits. In contrast, in macaques, the absence of HBZ was associated with reversion of the mutant virus to the wild-type genotype in 3 of the 4 animals within weeks from infection. Similarly, reversion to the wild type was observed in 2 of the 4 macaque inoculated with the p30 mutant. The 4 macaques exposed to the p12 knock remained seronegative, and only 2 animals were positive at a single time point for viral DNA in tissues. Interestingly, we found that the p12 and the p30 mutants were also severely impaired in their ability to replicate in human dendritic cells. These data suggest that infection of dendritic cells may be required for the establishment and maintenance of HTLV-1 infection in primate species.


Asunto(s)
Células Dendríticas/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Línea Celular , Cartilla de ADN/genética , ADN Viral/genética , Células Dendríticas/inmunología , Femenino , Eliminación de Gen , Genes Virales , Genes pX , Genotipo , Infecciones por HTLV-I/inmunología , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Técnicas In Vitro , Macaca mulatta , Mutagénesis , Mutación , Conejos , Especificidad de la Especie , Linfocitos T/inmunología , Linfocitos T/virología , Proteínas Virales/genética , Proteínas Virales/fisiología , Virulencia/genética , Virulencia/fisiología
12.
J Virol ; 83(22): 11673-81, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19726508

RESUMEN

The Vpu accessory gene that originated in the primate lentiviral lineage leading to human immunodeficiency virus type 1 is an antagonist of human tetherin/BST-2 restriction. Most other primate lentivirus lineages, including the lineage represented by simian immunodeficiency virus SIVagm from African green monkeys (AGMs), do not encode Vpu. While some primate lineages encode gene products other than Vpu that overcome tetherin/BST-2, we find that SIVagm does not antagonize physiologically relevant levels of AGM tetherin/BST-2. AGM tetherin/BST-2 can be induced by low levels of type I interferon and can potently restrict two independent strains of SIVagm. Although SIVagm Nef had an effect at low levels of AGM tetherin/BST-2, simian immunodeficiency virus SIVmus Vpu, from a virus that infects the related monkey Cercopithecus cephus, is able to antagonize even at high levels of AGM tetherin/BST-2 restriction. We propose that since the replication of SIVagm does not induce interferon production in vivo, tetherin/BST-2 is not induced, and therefore, SIVagm does not need Vpu. This suggests that primate lentiviruses evolve tetherin antagonists such as Vpu or Nef only if they encounter tetherin during the typical course of natural infection.


Asunto(s)
Chlorocebus aethiops/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Western Blotting , Línea Celular , Productos del Gen nef/fisiología , Interferón alfa-2 , Interferon beta-1b , Interferón-alfa/farmacología , Interferón beta/farmacología , Datos de Secuencia Molecular , Proteínas Recombinantes , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/fisiología
13.
EMBO Rep ; 10(2): 144-51, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19165139

RESUMEN

The retroviral integrase superfamily (RISF) comprises numerous important nucleic acid-processing enzymes, including transposases, integrases and various nucleases. These enzymes are involved in a wide range of processes such as transposition, replication and repair of DNA, homologous recombination, and RNA-mediated gene silencing. Two out of the four enzymes that are encoded by the human immunodeficiency virus--RNase H1 and integrase--are members of this superfamily. RISF enzymes act on various substrates, and yet show remarkable mechanistic and structural similarities. All share a common fold of the catalytic core and the active site, which is composed primarily of carboxylate residues. Here, I present RISF proteins from a structural perspective, describing the individual members and the common and divergent elements of their structures, as well as the mechanistic insights gained from the structures of RNase H1 enzyme complexes with RNA/DNA hybrids.


Asunto(s)
Integrasas/química , Familia de Multigenes , Proteínas de los Retroviridae/química , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Dominio Catalítico , Dimerización , Hidrólisis , Integrasas/fisiología , Mamíferos/metabolismo , Ratones , Modelos Moleculares , Ácidos Nucleicos/metabolismo , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas de los Retroviridae/fisiología , Ribonucleasa H/química , Ribonucleasa H/fisiología , Especificidad de la Especie , Relación Estructura-Actividad , Especificidad por Sustrato , Transposasas/química , Transposasas/fisiología , Proteínas Virales/química , Proteínas Virales/fisiología
14.
J Biol Chem ; 284(9): 5819-26, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19074429

RESUMEN

The APOBEC3 cytidine deaminases are part of the intrinsic defense of cells against retroviruses. Lentiviruses and spumaviruses have evolved essential accessory proteins, Vif and Bet, respectively, which counteract the APOBEC3 proteins. We show here that Bet of the Prototype foamy virus inhibits the antiviral APOBEC3C activity by a mechanism distinct to Vif: Bet forms a complex with APOBEC3C without inducing its degradation. Bet abolished APOBEC3C dimerization as shown by coimmunoprecipitation and cross-linking experiments. These findings implicate a physical interaction between Bet and the APOBEC3C. Subsequently, we identified the Bet interaction domain in human APOBEC3C in the predicted APOBEC3C dimerization site. Taken together, these data support the hypothesis that Bet inhibits incorporation of APOBEC3Cs into retroviral particles. Bet likely achieves this by trapping APOBEC3C protein in complexes rendering them unavailable for newly generated viruses due to direct immobilization.


Asunto(s)
Citidina Desaminasa/antagonistas & inhibidores , Citidina Desaminasa/metabolismo , Provirus/genética , Proteínas de los Retroviridae/fisiología , Desaminasa APOBEC-3G , Animales , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Reactivos de Enlaces Cruzados , Dimerización , Productos del Gen vif/fisiología , Humanos , Immunoblotting , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Macaca mulatta , Macaca nemestrina , Pan troglodytes , Virus Espumoso de los Simios , Transfección , Ensamble de Virus , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
15.
Cell Mol Life Sci ; 65(21): 3383-98, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18818872

RESUMEN

Up to 10% of the mouse genome is comprised of endogenous retrovirus (ERV) sequences, and most represent the remains of ancient germ line infections. Our knowledge of the three distinct classes of ERVs is inversely correlated with their copy number, and their characterization has benefited from the availability of divergent wild mouse species and subspecies, and from ongoing analysis of the Mus genome sequence. In contrast to human ERVs, which are nearly all extinct, active mouse ERVs can still be found in all three ERV classes. The distribution and diversity of ERVs has been shaped by host-virus interactions over the course of evolution, but ERVs have also been pivotal in shaping the mouse genome by altering host genes through insertional mutagenesis, by adding novel regulatory and coding sequences, and by their co-option by host cells as retroviral resistance genes. We review mechanisms by which an adaptive coexistence has evolved. (Part of a multi-author review).


Asunto(s)
Retrovirus Endógenos/fisiología , Interacciones Huésped-Patógeno/fisiología , Ratones/virología , Secuencia de Aminoácidos , Animales , Retrovirus Endógenos/clasificación , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidad , Evolución Molecular , Gammaretrovirus/clasificación , Gammaretrovirus/genética , Transferencia de Gen Horizontal , Genes de Partícula A Intracisternal/genética , Genoma , Interacciones Huésped-Patógeno/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis Insercional , Neoplasias/veterinaria , Neoplasias/virología , Receptores Virales/genética , Receptores Virales/fisiología , Retroelementos/genética , Retroelementos/fisiología , Infecciones por Retroviridae/veterinaria , Infecciones por Retroviridae/virología , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Enfermedades de los Roedores/virología , Infecciones Tumorales por Virus/veterinaria , Infecciones Tumorales por Virus/virología , Vertebrados/virología
16.
Annu Rev Genet ; 42: 709-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18694346

RESUMEN

For millions of years, retroviral infections have challenged vertebrates, occasionally leading to germline integration and inheritance as ERVs, genetic parasites whose remnants today constitute some 7% to 8% of the human genome. Although they have had significant evolutionary side effects, it is useful to view ERVs as fossil representatives of retroviruses extant at the time of their insertion into the germline and not as direct players in the evolutionary process itself. Expression of particular ERVs is associated with several positive physiological functions as well as certain diseases, although their roles in human disease as etiological agents, possible contributing factors, or disease markers-well demonstrated in animal models-remain to be established. Here we discuss ERV contributions to host genome structure and function, including their ability to mediate recombination, and physiological effects on the host transcriptome resulting from their integration, expression, and other events.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Retroviridae/patogenicidad , Empalme Alternativo , Animales , Evolución Biológica , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidad , Elementos de Facilitación Genéticos , Transferencia de Gen Horizontal , Humanos , Modelos Biológicos , Regiones Promotoras Genéticas , Retroviridae/genética , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología
17.
Apoptosis ; 13(6): 733-47, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18421579

RESUMEN

A universal cellular defense mechanism against viral invasion is the elimination of infected cells through apoptotic cell death. To counteract host defenses many viruses have evolved complex apoptosis evasion strategies. The oncogenic human retrovirus HTLV-1 is the etiological agent of adult-T-cell leukemia/lymphoma (ATLL) and the neurodegenerative disease known as HTLV-associated myelopathy/tropical spastic paraparesis (HAM/TSP). The poor prognosis in HTLV-1-induced ATLL is linked to the resistance of neoplastic T cells against conventional therapies and the immuno-compromised state of patients. Nevertheless, several studies have shown that the apoptotic pathway is largely intact and can be reactivated in ATLL tumor cells to induce specific killing. A better understanding of the molecular mechanisms employed by HTLV-1 to counteract cellular death pathways remains an important challenge for future therapies and the treatment of HTLV-1-associated diseases.


Asunto(s)
Apoptosis/fisiología , Transformación Celular Viral/fisiología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/virología , Apoptosis/efectos de los fármacos , Productos del Gen tax/fisiología , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Humanos , Quinasa I-kappa B/fisiología , Quinasas Janus/fisiología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Paraparesia Espástica Tropical/tratamiento farmacológico , Paraparesia Espástica Tropical/virología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptores de Interleucina-2/fisiología , Proteínas de los Retroviridae/fisiología , Factores de Transcripción STAT/fisiología , Tretinoina/farmacología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología
18.
Virology ; 364(2): 330-41, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17418360

RESUMEN

Although lentiviruses like HIV-1 are able to infect non-dividing cells, particular resting cells such as non-stimulated primary peripheral blood mononuclear cells (PBMC) are resistant to infection. In contrast to other lentiviruses, SIVsmmPBj can replicate in non-stimulated PBMC. Moreover, SIVsmmPBj-derived, but not HIV-1-derived, replication-incompetent vectors enable gene transfer into G(0)-arrested human cell lines and primary human monocytes. Here, we demonstrate that transduction of G(0)-arrested cell lines by SIVsmmPBj-derived vectors is independent of the viral accessory proteins Vif, Vpx, Vpr, or Nef. In contrast, for the transduction of primary human monocytes, the Vpx protein proved to be essential. However, trans-complementation of HIV-1 vectors with SIVsmmPBj Vpx did not provide the property of gene transfer into monocytes. Taken together, these data indicate that Vpx is essential for the infection of primary monocytes by SIVsmmPBj. Additionally, further genome functions besides the accessory proteins are required for the particular capacity of SIVsmmPBj in transduction or infection events.


Asunto(s)
Proteínas de los Retroviridae/fisiología , Virus de la Inmunodeficiencia de los Simios/fisiología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Proteínas Reguladoras y Accesorias Virales/fisiología , Secuencia de Bases , Línea Celular , ADN Viral/genética , Eliminación de Gen , Genes Virales , Prueba de Complementación Genética , Vectores Genéticos , VIH/genética , Humanos , Monocitos/virología , Fase de Descanso del Ciclo Celular , Proteínas de los Retroviridae/genética , Virus de la Inmunodeficiencia de los Simios/genética , Transducción Genética , Proteínas Reguladoras y Accesorias Virales/genética , Replicación Viral
19.
Virology ; 354(2): 225-39, 2006 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-16890266

RESUMEN

Human T-lymphotropic virus type-1 (HTLV-1) is a deltaretrovirus that causes adult T cell leukemia/lymphoma, and is implicated in a variety of lymphocyte-mediated inflammatory disorders. HTLV-1 provirus has regulatory and accessory genes in four pX open reading frames. HTLV-1 pX ORF-II encodes two proteins, p13II and p30II, which are incompletely defined in virus replication or pathogenesis. We have demonstrated that pX ORF-II mutations block virus replication in vivo and that ORF-II encoded p30II, a nuclear-localizing protein that binds with CREB-binding protein (CBP)/p300, represses CREB and Tax responsive element (TRE)-mediated transcription. Herein, we have identified p30II motifs important for p300 binding and in regulating TRE-mediated transcription in the absence and presence of HTLV-1 provirus. Within amino acids 100-179 of p30II, a region important for repression of LTR-mediated transcription, we identified a single lysine residue at amino acid 106 (K3) that significantly modulates the ability of p30II to repress TRE-mediated transcription. Exogenous p300, in a dose-responsive manner, reverses p30II-dependent repression of TRE-mediated transcription, in the absence or presence of the provirus, In contrast to wild type p300, p300 HAT mutants (defective in histone acetyltransferase activity) only partially rescued p30(II)-mediated LTR repression. Deacetylation by histone deacetylase-1 (HDAC-1) enhanced p30II-mediated LTR repression, while inhibition of deacetylation by trichostatin A decreases p30(II)-mediated LTR repression. Collectively, our data indicate that HTLV-1 p30II modulates viral gene expression in a cooperative manner with p300-mediated acetylation.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Histona Acetiltransferasas/fisiología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Proteínas de los Retroviridae/metabolismo , Secuencias Repetidas Terminales/fisiología , Factores de Transcripción/fisiología , Proteínas Virales/fisiología , Proteína de Unión a CREB/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación Viral de la Expresión Génica , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/virología , Transactivadores/genética , Transactivadores/metabolismo , Transcripción Genética/fisiología , Proteínas Virales/análisis , Factores de Transcripción p300-CBP
20.
J Biol Chem ; 281(33): 23414-24, 2006 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-16785240

RESUMEN

Whereas adaptive immunity has been extensively studied, very little is known about the innate immunity of the host to HTLV-I infection. HTLV-I-infected ATL patients have pronounced immunodeficiency associated with frequent opportunistic infections, and in these patients, concurrent infections with bacteria and/or parasites are known to increase risks of progression to ATL. The Toll-like receptor-4 (TLR4) activation in response to bacterial infection is essential for dendritic cell maturation and links the innate and adaptive immune responses. Recent reports indicate that TLR4 is targeted by viruses such as RSV, HCV, and MMTV. Here we report that HTLV-I has also evolved a protein that interferes with TLR4 signaling; p30 interacts with and inhibits the DNA binding and transcription activity of PU.1 resulting in the down-regulation of the TLR4 expression from the cell surface. Expression of p30 hampers the release of pro-inflammatory cytokines MCP-1, TNF-alpha, and IL-8 and stimulates release of anti-inflammatory IL-10 following stimulation of TLR4 in human macrophage. Finally, we found that p30 increases phosphorylation and inactivation of GSK3-beta a key step for IL-10 production. Our study suggests a novel function of p30, which may instigate immune tolerance by reducing activation of adaptive immunity in ATL patients.


Asunto(s)
Citocinas/metabolismo , Virus Linfotrópico T Tipo 1 Humano/química , Virus Linfotrópico T Tipo 1 Humano/fisiología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Proteínas de los Retroviridae/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Células COS , Proteína de Unión a CREB/fisiología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Humanos , Mediadores de Inflamación/fisiología , Macrófagos/virología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas de los Retroviridae/metabolismo , Receptor Toll-Like 4/fisiología , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Transactivadores/metabolismo , Técnicas del Sistema de Dos Híbridos , Factores de Transcripción p300-CBP/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...