Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Reprod Sci ; 28(7): 1850-1865, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33090377

RESUMO

Nlrp2 encodes a protein of the oocyte subcortical maternal complex (SCMC), required for embryo development. We previously showed that loss of maternal Nlrp2 in mice causes subfertility, smaller litters with birth defects, and growth abnormalities in offspring, indicating that Nlrp2 is a maternal effect gene and that all embryos from Nlrp2-deficient females that were cultured in vitro arrested before the blastocysts stage. Here, we used time-lapse microscopy to examine the development of cultured embryos from superovulated Nlrp2-deficient and wild-type mice after in vivo and in vitro fertilization. Embryos from Nlrp2-deficient females had similar abnormal cleavage and fragmentation and arrested by blastocyst stage, irrespective of fertilization mode. This indicates that in vitro fertilization does not further perturb or improve the development of cultured embryos. We also transferred embryos from superovulated Nlrp2-deficient and wild-type females to wild-type recipients to investigate if the abnormal reproductive outcomes of Nlrp2-deficient females are primarily driven by oocyte dysfunction or if a suboptimal intra-uterine milieu is a necessary factor. Pregnancies with transferred embryos from Nlrp2-deficient females produced smaller litters, stillbirths, and offspring with birth defects and growth abnormalities. This indicates that the reproductive phenotype is oocyte-specific and is not rescued by development in a wild-type uterus. We further found abnormal DNA methylation at two maternally imprinted loci in the kidney of surviving young adult offspring, confirming persistent DNA methylation disturbances in surviving offspring. These findings have implications for fertility treatments for women with mutations in NLRP2 and other genes encoding SCMC proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Desenvolvimento Embrionário/fisiologia , Fertilização in vitro , Oócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Blastocisto/metabolismo , Fragmentação do DNA , Metilação de DNA , Técnicas de Cultura Embrionária , Transferência Embrionária , Feminino , Camundongos , Gravidez , Resultado da Gravidez , Superovulação
2.
PLoS One ; 14(11): e0224876, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31703095

RESUMO

Gene-environment interactions contribute to the risk for Autism Spectrum Disorder (ASD). Among environmental factors, prenatal exposure to stress may increase the risk for ASD. To examine if there is an interaction between exposure to maternal stress and reduced dosage or loss of Shank3, wild-type (WT), heterozygous (HET) and homozygous (HOM) female mice carrying a deletion of exons four through nine of Shank3 (Shank3ex4-9) were exposed to chronic unpredictable mild stress (CUMS) from prior to conception throughout gestation. This study examined maternal care of these dams and the white matter microstructure in the brains of their adult male offspring. Overall, our findings suggest that maternal exposure to CUMS increased pup-directed care for dams of all three genotypes. Compared to WT and HET dams, HOM dams also exhibited increased maternal care behaviors with increased time spent in the nest and reduced cage exploration, regardless of exposure to CUMS. Diffusion tensor imaging showed higher mean fractional anisotropy in the hippocampal stratum radiatum of WT and HOM male offspring from dams exposed to CUMS and HOM offspring from unexposed dams, compared to WT male offspring from unexposed dams. These data support that CUMS in Shank3-mutant dams results in subtle maternal care alterations and long-lasting changes in the white matter of the hippocampus of their offspring.


Assuntos
Exposição Materna , Proteínas do Tecido Nervoso/genética , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico , Substância Branca/metabolismo , Substância Branca/fisiopatologia , Animais , Comportamento Animal , Imagem de Tensor de Difusão , Feminino , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Masculino , Comportamento Materno , Camundongos , Proteínas dos Microfilamentos , Mutação , Gravidez , Substância Branca/diagnóstico por imagem
3.
Prenat Diagn ; 39(4): 319-323, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30731021

RESUMO

OBJECTIVE: To examine factors that influence uptake of expanded carrier screening (ECS) among women undergoing preconception and prenatal genetic counseling. METHODS: We retrospectively reviewed 500 medical records from women with prenatal or preconception genetic counseling at a prenatal genetic counseling service. We tabulated acceptance of ECS by indication for genetic counseling along with demographic and pregnancy-related factors. RESULTS: ECS was offered to 483 of 500 women, and 192 (39.8%) accepted. Of the 67 women counseled preconceptionally, 46 (68.7%) accepted ECS. This was significantly more than for 416 women counseled during pregnancy, of whom 146 (35.1%) accepted (P ≤ 0.001). For pregnant patients, the mean gestational age of those accepting ECS (12 weeks 3 days; n = 146) was significantly lower than those declining (13 weeks 4 days; n = 270; P ≤ 0.001). The acceptance rates were 7 of 12 (58.3%, P = 0.195) for Ashkenazi Jewish women, 12 of 41 (29.3%; P = 0.186) for Asian women, and 7 of 25 (28.0%; P = 0.241) for women of mixed ethnicity. CONCLUSIONS: These results suggest that receiving genetic counseling prior to or earlier in the first trimester is associated with acceptance of ECS and support the importance of early genetic counseling about carrier screening options.


Assuntos
Triagem de Portadores Genéticos/métodos , Aconselhamento Genético/estatística & dados numéricos , Participação do Paciente/estatística & dados numéricos , Primeiro Trimestre da Gravidez , Diagnóstico Pré-Natal , Adulto , Etnicidade/estatística & dados numéricos , Feminino , Triagem de Portadores Genéticos/estatística & dados numéricos , Aconselhamento Genético/métodos , Idade Gestacional , Humanos , Masculino , Programas de Rastreamento/métodos , Programas de Rastreamento/estatística & dados numéricos , Gravidez , Cuidado Pré-Natal/métodos , Cuidado Pré-Natal/estatística & dados numéricos , Diagnóstico Pré-Natal/métodos , Diagnóstico Pré-Natal/estatística & dados numéricos , Estudos Retrospectivos , Adulto Jovem
5.
PLoS One ; 12(1): e0170127, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28099477

RESUMO

Offspring of murine dams chronically fed a protein-restricted diet have an increased risk for metabolic and neurobehavioral disorders. Previously we showed that adult offspring, developmentally exposed to a chronic maternal low-protein (MLP) diet, had lower body and hind-leg muscle weights and decreased liver enzyme serum levels. We conducted energy expenditure, neurobehavioral and circadian rhythm assays in male offspring to examine mechanisms for the body-weight phenotype and assess neurodevelopmental implications of MLP exposure. C57BL/6J dams were fed a protein restricted (8%protein, MLP) or a control protein (20% protein, C) diet from four weeks before mating until weaning of offspring. Male offspring were weaned to standard rodent diet (20% protein) and single-housed until 8-12 weeks of age. We examined body composition, food intake, energy expenditure, spontaneous rearing activity and sleep patterns and performed behavioral assays for anxiety (open field activity, elevated plus maze [EPM], light/dark exploration), depression (tail suspension and forced swim test), sociability (three-chamber), repetitive (marble burying), learning and memory (fear conditioning), and circadian behavior (wheel-running activity during light-dark and constant dark cycles). We also measured circadian gene expression in hypothalamus and liver at different Zeitgeber times (ZT). Male offspring from separate MLP exposed dams had significantly greater body fat (P = 0.03), less energy expenditure (P = 0.004), less rearing activity (P = 0.04) and a greater number of night-time rest/sleep bouts (P = 0.03) compared to control. MLP offspring displayed greater anxiety-like behavior in the EPM (P<0.01) but had no learning and memory deficit in fear-conditioning assay (P = 0.02). There was an effect of time on Per1, Per 2 and Clock circadian gene expression in the hypothalamus but not on circadian behavior. Thus, transplacental and early developmental exposure of dams to chronic MLP reduces food intake and energy expenditure, increases anxiety like behavior and disturbs sleep patterns but not circadian rhythm in adult male offspring.


Assuntos
Ansiedade/etiologia , Ritmo Circadiano/fisiologia , Dieta com Restrição de Proteínas/efeitos adversos , Metabolismo Energético , Sono/fisiologia , Tecido Adiposo , Animais , Comportamento Animal , Ritmo Circadiano/genética , Feminino , Expressão Gênica , Hipotálamo/fisiologia , Fígado/fisiologia , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Camundongos Endogâmicos C57BL
6.
AIDS Res Hum Retroviruses ; 31(1): 137-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24528253

RESUMO

The histone deacetylase inhibitor (HDACi) suberoylanilide hydroxyamic acid (SAHA), also known as vorinostat, has recently been reported to activate latent HIV-1 in patients undergoing antiretroviral therapy. It is possible that SAHA reactivation of latent viruses may involve effects on cellular transcription factors such as positive transcription elongation factor b (P-TEFb), a protein kinase whose core is composed of CDK9 and Cyclin T1. P-TEFb is recruited by the HIV-1 Tat protein to activate productive RNA polymerase II elongation of the integrated provirus. We found that SAHA treatment of isolated resting CD4(+) T cells induced CDK9 Thr-186 (T-loop) phosphorylation in six of eight healthy donors and increased Cyclin T1 expression in one donor; Thr-186 phosphorylation is required for P-TEFb function. Disulfiram, another small molecule currently under evaluation in clinical trials for reactivation of latent HIV-1, was also found capable of inducing CDK9 Thr-186 phosphorylation and Cyclin T1 levels in resting CD4(+) T cells from healthy donors. In a Jurkat CD4(+) T cells HIV-1 latency system, disulfiram reactivated the latent provirus and induced CDK9 Thr-186 phosphorylation. Our findings suggest that small molecules capable of reactivating latent HIV-1 in resting CD4(+) T cells may function in part by increasing CDK9 Thr-186 phosphorylation and perhaps Cyclin T1 expression, thereby up-regulating P-TEFb function.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quinase 9 Dependente de Ciclina/metabolismo , Dissulfiram/farmacologia , Ácidos Hidroxâmicos/farmacologia , Fator B de Elongação Transcricional Positiva/metabolismo , Inibidores de Acetaldeído Desidrogenases/farmacologia , Linhagem Celular , Ciclina T/biossíntese , Infecções por HIV/virologia , HIV-1/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Células Jurkat , Fosforilação/efeitos dos fármacos , Ativação Viral/efeitos dos fármacos , Latência Viral , Vorinostat
7.
Bioorg Med Chem ; 22(21): 6112-6, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25260957

RESUMO

Screening of a small library of natural product extracts derived from endophytic fungi of the Sonoran desert plants in a cell-based anti-HIV assay involving T-cells infected with the HIV-1 virus identified the EtOAc extract of a fermentation broth of Alternaria tenuissima QUE1Se inhabiting the stem tissue of Quercus emoryi as a promising candidate for further investigation. Bioactivity-guided fractionation of this extract led to the isolation and identification of two new metabolites, altertoxins V (1) and VI (2) together with the known compounds, altertoxins I (3), II (4), and III (5). The structures of 1 and 2 were determined by detailed spectroscopic analysis and those of 3-5 were established by comparison with reported data. When tested in our cell-based assay at concentrations insignificantly toxic to T-cells, altertoxins V (1), I (3), II (4), and III (5) completely inhibited replication of the HIV-1 virus at concentrations of 0.50, 2.20, 0.30, and 1.50 µM, respectively. Our findings suggest that the epoxyperylene structural scaffold in altertoxins may be manipulated to produce potent anti-HIV therapeutics.


Assuntos
Alternaria/química , Fármacos Anti-HIV/farmacologia , Benzo(a)Antracenos/farmacologia , HIV-1/efeitos dos fármacos , Perileno/análogos & derivados , Alternaria/fisiologia , Fármacos Anti-HIV/química , Fármacos Anti-HIV/isolamento & purificação , Benzo(a)Antracenos/química , Benzo(a)Antracenos/isolamento & purificação , Endófitos , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Humanos , Perileno/química , Perileno/isolamento & purificação , Perileno/farmacologia , Quercus/fisiologia , Linfócitos T/virologia
8.
Open Virol J ; 7: 72-80, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23961302

RESUMO

Most antiretroviral drugs currently in use to treat an HIV-1 infection are chemically synthesized and lead to the development of viral resistance, as well as cause severe toxicities. However, a largely unexplored source for HIV-1 drug discovery is endophytic fungi that live in a symbiotic relationship with plants. These fungi produce biologically active secondary metabolites, which are natural products that are beneficial to the host. We prepared several hundred extracts from endophytic fungi of desert plants and evaluated the inhibitory effects on HIV-1 replication of those extracts that showed less than 30% cytotoxicity in T-lymphocytes. Those extracts that inhibited viral replication were fractionated in order to isolate the compounds responsible for activity. Multiple rounds of fractionation and antiviral evaluation lead to the identification of four compounds, which almost completely impede HIV-1 replication. These studies demonstrate that metabolites from endophytic fungi of desert plants can serve as a viable source for identifying potent inhibitors of HIV-1 replication.

9.
Retrovirology ; 9: 90, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23110726

RESUMO

BACKGROUND: HIV-1 Tat activates RNA Polymerase II (RNAP II) elongation of the integrated provirus by recruiting a protein kinase known as P-TEFb to TAR RNA at the 5' end of nascent viral transcripts. The catalytic core of P-TEFb contains CDK9 and Cyclin T1 (CCNT1). A human endogenous complexome has recently been described - the set of multi-protein complexes in HeLa cell nuclei. We mined this complexome data set and identified 12 distinct multi-protein complexes that contain both CDK9 and CCNT1. We have termed these complexes CCAPs for CDK9/CCNT1-associated protein complexes. Nine CCAPs are novel, while three were previously identified as Core P-TEFb, the 7SK snRNP, and the Super-Elongation Complex. We have investigated the role of five newly identified CCAPs in Tat function and viral gene expression. RESULTS: We examined five CCAPs that contain: 1) PPP1R10/TOX3/WDR82; 2) TTF2; 3) TPR; 4) WRNIP1; 5) FBXO11/CUL1/SKP1. SiRNA depletions of protein subunits of the five CCAPs enhanced Tat activation of an integrated HIV-1 LTR-Luciferase reporter in TZM-bl cells. Using plasmid transfection assays in HeLa cells, we also found that siRNA depletions of TTF2, FBXO11, PPP1R10, WDR82, and TOX3 enhanced Tat activation of an HIV-1 LTR-luciferase reporter, but the depletions did not enhance expression of an NF-κB reporter plasmid with the exception of PPP1R10. We found no evidence that depletion of CCAPs perturbed the level of CDK9/CCNT1 in the 7SK snRNP. We also found that the combination of siRNA depletions of both TTF2 and FBXO11 sensitized a latent provirus in Jurkat cells to reactivation by sub-optimal amounts of αCD3/CD28 antibodies. CONCLUSIONS: Our results identified five novel CDK9/CCNT1 complexes that are capable of negative regulation of HIV-1 Tat function and viral gene expression. Because siRNA depletions of CCAPs enhance Tat function, it is possible that these complexes reduce the level of CDK9 and CCNT1 available for Tat, similar to the negative regulation of Tat by the 7SK snRNP. Our results highlight the complexity in the biological functions of CDK9 and CCNT1.


Assuntos
Ciclina T/metabolismo , Quinase 9 Dependente de Ciclina/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Complexos Multiproteicos/metabolismo , RNA Interferente Pequeno/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Transcrição Gênica , Fatores de Virulência/metabolismo
10.
Retrovirology ; 9: 52, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22727189

RESUMO

BACKGROUND: Processive elongation of the integrated HIV-1 provirus is dependent on recruitment of P-TEFb by the viral Tat protein to the viral TAR RNA element. P-TEFb kinase activity requires phosphorylation of Thr186 in the T-loop of the CDK9 subunit. In resting CD4+T cells, low levels of T-loop phosphorylated CDK9 are found, which increase significantly upon activation. This suggests that the phosphorylation status of the T-loop is actively regulated through the concerted actions of cellular proteins such as Ser/Thr phosphatases. We investigated the role of phosphatase PPM1A in regulating CDK9 T-loop phosphorylation and its effect on HIV-1 proviral transcription. RESULTS: We found that overexpression of PPM1A inhibits HIV-1 gene expression during viral infection and this required PPM1A catalytic function. Using an artificial CDK tethering system, we further found that PPM1A inhibits CDK9, but not CDK8 mediated activation of the HIV-1 LTR. SiRNA depletion of PPM1A in resting CD4+T cells increased the level of CDK9 T-loop phosphorylation and enhanced HIV-1 gene expression. We also observed that PPM1A protein levels are relatively high in resting CD4+T cells and are not up-regulated upon T cell activation. CONCLUSIONS: Our results establish a functional link between HIV-1 replication and modulation of CDK9 T-loop phosphorylation by PPM1A. PPM1A represses HIV-1 gene expression by inhibiting CDK9 T-loop phosphorylation, thus reducing the amount of active P-TEFb available for recruitment to the viral LTR. We also infer that PPM1A enzymatic activity in resting and activated CD4+ T cells are likely regulated by as yet undefined factors.


Assuntos
Linfócitos T CD4-Positivos/virologia , Regulação Viral da Expressão Gênica , HIV-1/genética , Fosfoproteínas Fosfatases/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Quinase 8 Dependente de Ciclina/genética , Quinase 8 Dependente de Ciclina/metabolismo , Quinase 9 Dependente de Ciclina/genética , Quinase 9 Dependente de Ciclina/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Infecções por HIV/virologia , Repetição Terminal Longa de HIV , HIV-1/metabolismo , Células HeLa , Humanos , Fosfoproteínas Fosfatases/genética , Fosforilação , Fator B de Elongação Transcricional Positiva/genética , Proteína Fosfatase 2C , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transfecção
11.
J Cell Physiol ; 227(2): 609-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21448926

RESUMO

Eukaryotic RNA polymerase II transcriptional elongation is a tightly regulated process and is dependent upon positive transcription elongation factor-b (P-TEFb). The core P-TEFb complex is composed of Cdk9 and Cyclin T and is essential for the expression of most protein coding genes. Cdk9 kinase function is dependent upon phosphorylation of Thr186 in its T-loop. In this study, we examined kinases and signaling pathways that influence Cdk9 T-loop phosphorylation. Using an RNAi screen in HeLa cells, we found that Cdk9 T-loop phosphorylation is regulated by Ca(2+)/calmodulin-dependent kinase 1D (CaMK1D). Using small molecules inhibitors in HeLa cells and primary CD4(+) T lymphocytes, we found that the Ca(2+) signaling pathway is required for Cdk9 T-loop phosphorylation. Inhibition of Ca(2+) signaling led to dephosphorylation of Thr186 on Cdk9. In reporter plasmid assays, inhibition of the Ca(2+) signaling pathway repressed the PCNA promoter and HIV-1 Tat transactivation of the HIV-1 LTR, but not HTLV-1 Tax transactivation of the HTLV-1 LTR, suggesting that perturbation of the Ca(2+) pathway and reduction of Cdk9 T-loop phosphorylation inhibits transcription units that have a rigorous requirement for P-TEFb function.


Assuntos
Sinalização do Cálcio/fisiologia , Quinase 9 Dependente de Ciclina/metabolismo , Linfócitos T CD4-Positivos/fisiologia , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/metabolismo , Quinase 9 Dependente de Ciclina/química , Quinase 9 Dependente de Ciclina/genética , Regulação da Expressão Gênica/fisiologia , Células HeLa , Humanos , Fator B de Elongação Transcricional Positiva/metabolismo , Complexo de Endopeptidases do Proteassoma , Interferência de RNA , Transdução de Sinais/fisiologia
12.
Biology (Basel) ; 1(1): 94-115, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24832049

RESUMO

Productive transcription of the integrated HIV-1 provirus is restricted by cellular factors that inhibit RNA polymerase II elongation. The viral Tat protein overcomes this by recruiting a general elongation factor, P-TEFb, to the TAR RNA element that forms at the 5' end of nascent viral transcripts. P-TEFb exists in multiple complexes in cells, and its core consists of a kinase, Cdk9, and a regulatory subunit, either Cyclin T1 or Cyclin T2. Tat binds directly to Cyclin T1 and thereby targets the Cyclin T1/P-TEFb complex that phosphorylates the CTD of RNA polymerase II and the negative factors that inhibit elongation, resulting in efficient transcriptional elongation. P-TEFb is tightly regulated in cells infected by HIV-1-CD4+ T lymphocytes and monocytes/macrophages. A number of mechanisms have been identified that inhibit P-TEFb in resting CD4+ T lymphocytes and monocytes, including miRNAs that repress Cyclin T1 protein expression and dephosphorylation of residue Thr186 in the Cdk9 T-loop. These repressive mechanisms are overcome upon T cell activation and macrophage differentiation when the permissivity for HIV-1 replication is greatly increased. This review will summarize what is currently known about mechanisms that regulate P-TEFb and how this regulation impacts HIV-1 replication and latency.

13.
PLoS One ; 6(11): e27271, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22102884

RESUMO

Nasal NK/T cell lymphomas (NKTCL) are a subset of aggressive Epstein-Barr virus (EBV)-associated non-Hodgkin's lymphomas. The role of EBV in pathogenesis of NKTCL is not clear. Intriguingly, EBV encodes more than 40 microRNAs (miRNA) that are differentially expressed and largely conserved in lymphocryptoviruses. While miRNAs play a critical role in the pathogenesis of cancer, especially lymphomas, the expression and function of EBV transcribed miRNAs in NKTCL are not known. To examine the role of EBV miRNAs in NKTCL, we used microarray profiling and qRT-PCR to identify and validate expression of viral miRNAs in SNK6 and SNT16 cells, which are two independently derived NKTCL cell lines that maintain the type II EBV latency program. All EBV BART miRNAs except BHRF-derived miRNAs were expressed and some of these miRNAs are expressed at higher levels than in nasopharyngeal carcinomas. Modulating the expression of BART9 with antisense RNAs consistently reduced SNK6 and SNT16 proliferation, while antisense RNAs to BARTs-7 and -17-5p affected proliferation only in SNK6 cells. Furthermore, the EBV LMP-1 oncoprotein and transcript levels were repressed when an inhibitor of BART9 miRNA was transfected into SNK6 cells, and overexpression of BART9 miRNA increased LMP-1 protein and mRNA expression. Our data indicate that BART9 is involved in NKTCL proliferation, and one of its mechanisms of action appears to be regulating LMP-1 levels. Our findings may have direct application for improving NKTCL diagnosis and for developing possible novel treatment approaches for this tumor, for which current chemotherapeutic drugs have limited effectiveness.


Assuntos
Infecções por Vírus Epstein-Barr/patologia , Células Matadoras Naturais/patologia , Linfoma de Células T/patologia , MicroRNAs/genética , Mucosa Nasal/patologia , Neoplasias Nasofaríngeas/patologia , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma , Proliferação de Células , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Perfilação da Expressão Gênica , Herpesvirus Humano 4/genética , Humanos , Hibridização In Situ , Células Matadoras Naturais/metabolismo , Linfoma de Células T/metabolismo , Linfoma de Células T/virologia , Mucosa Nasal/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas da Matriz Viral/genética , Proteínas Virais/metabolismo , Latência Viral , Replicação Viral
14.
BMC Res Notes ; 4: 260, 2011 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-21791050

RESUMO

BACKGROUND: The elongation phase, like other steps of transcription by RNA Polymerase II, is subject to regulation. The positive transcription elongation factor b (P-TEFb) complex allows for the transition of mRNA synthesis to the productive elongation phase. P-TEFb contains Cdk9 (Cyclin-dependent kinase 9) as its catalytic subunit and is regulated by its Cyclin partners, Cyclin T1 and Cyclin T2. The HIV-1 Tat transactivator protein enhances viral gene expression by exclusively recruiting the Cdk9-Cyclin T1 P-TEFb complex to a RNA element in nascent viral transcripts called TAR. The expression patterns of Cyclin T1 and Cyclin T2 in primary monocytes and CD4+ T cells suggests that Cyclin T2 may be generally involved in expression of constitutively expressed genes in quiescent cells, while Cyclin T1 may be involved in expression of genes up-regulated during macrophage differentiation, T cell activation, and conditions of increased metabolic activity To investigate this issue, we wished to identify the sets of genes whose levels are regulated by either Cyclin T2 or Cyclin T1. FINDINGS: We used shRNA lentiviral vectors to stably deplete either Cyclin T2 or Cyclin T1 in HeLa cells. Total RNA extracted from these cells was subjected to cDNA microarray analysis. We found that 292 genes were down- regulated by depletion of Cyclin T2 and 631 genes were down-regulated by depletion of Cyclin T1 compared to cells transduced with a control lentivirus. Expression of 100 genes was commonly reduced in either knockdown. Additionally, 111 and 287 genes were up-regulated when either Cyclin T2 or Cyclin T1 was depleted, respectively, with 45 genes in common. CONCLUSIONS: These results suggest that there is limited redundancy in genes regulated by Cyclin T1 or Cyclin T2.

15.
J Leukoc Biol ; 86(6): 1345-50, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19741158

RESUMO

The cellular kinase complex P-TEFb is composed of Cdk9 and cyclin T, and it is required for expression of most protein-coding genes by RNAP II. Cdk9 has been shown recently to be activated in cis by autophosphorylation of Thr186 in its T-loop. Using a phosphospecific Cdk9 antibody, we examined the level of Cdk9 T-loop phosphorylation in resting and activated CD4(+) T lymphocytes. Cdk9 T-loop phosphorylation was found to be low-to-undetectable in resting CD4(+) T lymphocytes, and upon activation by distinct stimuli, there is a rapid (<1 h) increase in pCdk9 that does not require protein synthesis. The low level of Cdk9 T-loop phosphorylation was not to be a result of the absence of an associated regulatory cyclin partner. These observations suggest that autophosphorylation of the Cdk9 T-loop is repressed in resting CD4(+) T lymphocytes. The low level of T-loop phosphorylation in resting cells is also reflected in a low level of phosphorylation of Ser2 in the carboxyl terminal domain of RNAP II, suggesting that lack of Cdk9 T-loop autophosphorylation may limit RNAP II elongation in quiescent CD4(+) T lymphocytes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quinase 9 Dependente de Ciclina/imunologia , Regulação da Expressão Gênica/imunologia , Ativação Linfocitária/imunologia , RNA Polimerase II/imunologia , Linfócitos T CD4-Positivos/enzimologia , Ciclina T/genética , Ciclina T/metabolismo , Quinase 9 Dependente de Ciclina/metabolismo , Humanos , Fosforilação/imunologia , Fator B de Elongação Transcricional Positiva/genética , Fator B de Elongação Transcricional Positiva/metabolismo , Estrutura Terciária de Proteína/fisiologia , RNA Polimerase II/metabolismo
16.
Virology ; 385(1): 28-38, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19100594

RESUMO

We have recently shown an increased HIV-1 replication and gene expression in neonatal (cord) blood mononuclear cells compared with adult cells, which could be due to HIV-1 integration as it targets active host genes. Here we have characterized 468 HIV-1 integration sites within cord and adult blood T-lymphocytes and monocyte-derived macrophages (MDM) from five donors. Several functional classes of genes were identified by gene ontology to be over represented, including genes for cellular components, maintenance of intracellular environment, enzyme regulation, cellular metabolism, catalytic activity and cation transport. Numerous potential transcription factor binding sites at the sites of integration were identified. Furthermore, the genes at the site of integration, transcription factors which potentially bind upstream of the HIV-1 promoter and factors that assist HIV-1 integration were found to be expressed at higher levels in cord than adult cells. Taken together, these results suggest HIV-1 integration occurred in a more actively transcribed genes in neonatal cells compared with adult cells, which may help explain a higher level of HIV-1 gene expression and replication in neonatal compared with adult cells.


Assuntos
Infecções por HIV/virologia , HIV-1/fisiologia , Leucócitos Mononucleares/virologia , RNA Viral/metabolismo , Integração Viral , Adulto , Células Sanguíneas/metabolismo , Células Sanguíneas/virologia , Sangue Fetal/imunologia , Sangue Fetal/metabolismo , Sangue Fetal/virologia , Regulação Viral da Expressão Gênica , Genes Virais/fisiologia , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/genética , Humanos , Recém-Nascido/imunologia , Recém-Nascido/metabolismo , Leucócitos Mononucleares/metabolismo , RNA Viral/genética , Replicação Viral
17.
J Biol Chem ; 283(48): 33578-84, 2008 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-18829461

RESUMO

Cdk9 is the catalytic subunit of a general RNA polymerase II elongation factor known as positive transcription elongation factor b (P-TEFb). The kinase function of P-TEFb requires phosphorylation of Thr-186 in the T-loop of Cdk9 to allow substrates to access the catalytic core of the enzyme. To identify human phosphatases that dephosphorylate the T-loop of Cdk9, we used a Thr-186-phosphospecific antiserum to screen a phosphatase expression library. Overexpression of PPM1A and the related PPM1B greatly reduced Cdk9 T-loop phosphorylation in vivo. PPM1A and Cdk9 appear to associate in vivo as the proteins could be co-immunoprecipitated. The short hairpin RNA depletion of PPM1A resulted in an increase in Cdk9 T-loop phosphorylation. In phosphatase reactions in vitro, purified PPM1A could dephosphorylate Thr-186 both with and without the association of 7SK RNA, a small nuclear RNA that is bound to approximately 50% of total cellular P-TEFb. PPM1B only efficiently dephosphorylated Cdk9 Thr-186 in vitro when 7SK RNA was depleted from P-TEFb. Taken together, our data indicate that PPM1A and to some extent PPM1B are important negative regulators of P-TEFb function.


Assuntos
Quinase 9 Dependente de Ciclina/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Domínio Catalítico/fisiologia , Quinase 9 Dependente de Ciclina/genética , Células HeLa , Humanos , Fosfoproteínas Fosfatases/genética , Fosforilação/fisiologia , Fator B de Elongação Transcricional Positiva/genética , Proteína Fosfatase 2C , Estrutura Secundária de Proteína/fisiologia , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo
18.
PLoS One ; 3(9): e3146, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18773076

RESUMO

HIV-1 is dependent upon cellular co-factors to mediate its replication cycle in CD4(+) T cells and macrophages, the two major cell types infected by the virus in vivo. One critical co-factor is Cyclin T1, a subunit of a general RNA polymerase II elongation factor known as P-TEFb. Cyclin T1 is targeted directly by the viral Tat protein to activate proviral transcription. Cyclin T1 is up-regulated when resting CD4(+) T cells are activated and during macrophage differentiation or activation, conditions that are also necessary for high levels of HIV-1 replication. Because Cyclin T1 is a subunit of a transcription factor, the up-regulation of Cyclin T1 in these cells results in the induction of cellular genes, some of which might be HIV-1 co-factors. Using shRNA depletions of Cyclin T1 and transcriptional profiling, we identified 54 cellular mRNAs that appear to be Cyclin T1-dependent for their induction in activated CD4(+) T Jurkat T cells and during differentiation and activation of MM6 cells, a human monocytic cell line. The promoters for these Cyclin T1-dependent genes (CTDGs) are over-represented in two transcription factor binding sites, SREBP1 and ARP1. Notably, 10 of these CTDGs have been reported to be involved in HIV-1 replication, a significant over-representation of such genes when compared to randomly generated lists of 54 genes (p value<0.00021). The results of siRNA depletion and dominant-negative protein experiments with two CTDGs identified here, CDK11 and Casein kinase 1 gamma 1, suggest that these genes are involved either directly or indirectly in HIV-1 replication. It is likely that the 54 CTDGs identified here include novel HIV-1 co-factors. The presence of CTDGs in the protein space that was available for HIV-1 to sample during its evolution and acquisition of Tat function may provide an explanation for why CTDGs are enriched in viral co-factors.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/virologia , Ciclinas/metabolismo , Regulação Viral da Expressão Gênica , HIV-1/metabolismo , Macrófagos/citologia , Macrófagos/virologia , Sítios de Ligação , Caseína Quinase I/metabolismo , Ciclina T , Quinases Ciclina-Dependentes/metabolismo , Genes Dominantes , Humanos , Células Jurkat , Ativação Linfocitária , Monócitos/citologia , Linfócitos T/citologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
19.
AIDS Res Hum Retroviruses ; 24(3): 437-45, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18327987

RESUMO

Human immunodeficiency virus type 1 (HIV-1) long terminal repeat (LTR) sequences were characterized from six mother-infant pairs following vertical transmission. The LTR sequences exhibited a low degree of heterogeneity within mothers, within infants, and between epidemiologically linked mother-infant pairs. However, LTR sequences were more heterogeneous between epidemiologically unlinked individuals compared with linked mother-infant pairs. These data were further supported by low estimates of genetic diversity and clustering of each mother-infant pair's sequences into a separate subtree as well as the presence of common signature sequences between mother-infant pairs. The functional domains essential for LTR (promoter) function, including the promoter (TATAA), enhancers (three Sp-I and two NF-kappaB), the modulatory regions (two AP-I sites, two NFAT, one NF-IL6 site, one Ets-1, and one USF-1), and the TAR region were generally conserved among mother-infant pairs. Taken together, limited heterogeneity and conservation of functional domains in the LTR following vertical transmission support the notion that a functional LTR is critical in viral replication and pathogenesis in HIV-1-infected mothers and their infected infants.


Assuntos
Infecções por HIV/transmissão , Infecções por HIV/virologia , HIV-1/genética , Transmissão Vertical de Doenças Infecciosas , Sequências Repetidas Terminais/genética , Adulto , Pré-Escolar , Clonagem Molecular , Análise por Conglomerados , Sequência Conservada , Feminino , HIV-1/isolamento & purificação , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Mães , Polimorfismo Genético , RNA Viral/genética , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico
20.
Virol J ; 4: 126, 2007 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-18036244

RESUMO

BACKGROUND: Several subtypes of HIV-1 circulate in infected people worldwide, including subtype B in the United States and subtype C in Africa and India. To understand the biological properties of HIV-1 subtype C, including cellular tropism, virus entry, replication efficiency and cytopathic effects, we reciprocally inserted our previously characterized envelope V3-V5 regions derived from 9 subtype C infected patients from India into a subtype B molecular clone, pNL4-3. Equal amounts of the chimeric viruses were used to infect T-lymphocyte cell lines (A3.01 and MT-2), coreceptor cell lines (U373-MAGI-CCR5/CXCR4), primary blood T-lymphocytes (PBL) and monocyte-derived macrophages (MDM). RESULTS: We found that subtype C envelope V3-V5 region chimeras failed to replicate in T-lymphocyte cell lines but replicated in PBL and MDM. In addition, these chimeras were able to infect U373MAGI-CD4+-CCR5+ but not U373MAGI-CD4+-CXCR4+ cell line, suggesting CCR5 coreceptor utilization and R5 phenotypes. These subtype C chimeras were unable to induce syncytia in MT-2 cells, indicative of non-syncytium inducing (NSI) phenotypes. More importantly, the subtype C envelope chimeras replicated at higher levels in PBL and MDM compared with subtype B chimeras and isolates. Furthermore, the higher levels subtype C chimeras replication in PBL and MDM correlated with increased virus entry in U373MAGI-CD4+-CCR5+. CONCLUSION: Taken together, these results suggest that the envelope V3 to V5 regions of subtype C contributed to higher levels of HIV-1 replication compared with subtype B chimeras, which may contribute to higher viral loads and faster disease progression in subtype C infected individuals than other subtypes as well as rapid HIV-1 subtype C spread in India.


Assuntos
Proteína gp120 do Envelope de HIV/fisiologia , Infecções por HIV/virologia , HIV-1/fisiologia , Macrófagos/virologia , Receptores CCR5/fisiologia , Receptores CXCR4/fisiologia , Linfócitos T/virologia , Adolescente , Adulto , Sequência de Aminoácidos , Linhagem Celular , Feminino , Células Gigantes/imunologia , Células Gigantes/virologia , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/sangue , Infecções por HIV/imunologia , HIV-1/genética , HIV-1/imunologia , HIV-1/metabolismo , Células HeLa , Humanos , Macrófagos/imunologia , Masculino , Dados de Sequência Molecular , Receptores CCR5/imunologia , Receptores CCR5/metabolismo , Receptores CXCR4/imunologia , Receptores CXCR4/metabolismo , Proteínas Recombinantes de Fusão/genética , Linfócitos T/imunologia , Internalização do Vírus , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...