Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Biochem ; 118(11): 4020-4032, 2017 11.
Article in English | MEDLINE | ID: mdl-28422315

ABSTRACT

Positive transcription elongation factor-b (P-TEFb) is required for the release of RNA polymerase II (RNAPII) from its pause near the gene promoters and thus for efficient proceeding to the transcription elongation. It consists of two core subunits-CDK9 and one of T-typed or K-typed cyclin, of which, cyclin T1/CDK9 is the major and most studied combination. We have previously identified a novel splice variant of cyclin T1, cyclin T1b, which negatively regulates the transcription elongation of HIV-1 genes as well as several host genes. In this study, we revealed the serine-arginine-rich protein, ASF/SF2, as a regulatory factor of the alternative splicing of cyclin T1 gene. ASF/SF2 promotes the production of cyclin T1b versus cyclin T1a and regulates the expression of cyclin T1-depedent genes at the transcription level. We further found that a cis-element on exon 8 is responsible for the skipping of exon 7 mediated by ASF/SF2. Collectively, ASF/SF2 is identified as a splicing regulator of cyclin T1, which contributes to the control of the subsequent transcription events. J. Cell. Biochem. 118: 4020-4032, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Alternative Splicing/physiology , Cyclin T/biosynthesis , Serine-Arginine Splicing Factors/metabolism , Cell Line , Cyclin T/genetics , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Serine-Arginine Splicing Factors/genetics
2.
Retrovirology ; 12: 36, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25928027

ABSTRACT

BACKGROUND: As a member of the tumor necrosis factor receptor (TNFR) protein superfamily, equine lentivirus receptor 1 (ELR1) has been shown to be expressed in various equine cells that are permissive for equine infectious anemia virus (EIAV) replication. The EIAV Tat protein (eTat) activates transcription initiated at the viral long terminal repeat (LTR) promoter through a unique mechanism that requires the recruitment of the equine cyclin T1 (eCT1) cofactor into the viral TAR RNA target element. In vitro studies have demonstrated that mouse fibroblast cell lines (e.g., NIH 3T3 cells) that express the EIAV receptor ELR1 and eCT1 support the productive replication of EIAV. Therefore, we constructed transgenic eCT1- and ELR1-expressing mice to examine whether they support in vivo EIAV replication. FINDINGS: For the first time, we constructed mice transgenic for ELR1 and eCT1. Real-time reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis confirmed that ELR1 and eCT1 were expressed in the transgenic mouse tissues, particularly in the intestines, spleen and lymph nodes. Consistent with the results of EIAV infection in NIH 3T3 cells expressing ELR1 and eCT1, mouse embryonic fibroblasts (MEFs) from the transgenic mice could support EIAV replication. More importantly, this virus could infect and replicate in mouse blood monocyte-derived macrophages (mMDMs). Macrophages are the principle target cell of EIAV in its natural hosts. Furthermore, after the transgenic mice were inoculated with EIAV, the virus could be detected not only in the plasma of the circulating blood but also in multiple organs, among which, the spleen and lymph nodes were the predominant sites of EIAV replication. Finally, we found that consistent with high viral replication levels, the relevant pathological changes occurred in the spleen and lymph nodes. CONCLUSIONS: Our results show that mice transgenic for ELR1 and eCT1 are susceptible to EIAV infection and replication. Further, EIAV infection can cause lesions on the spleen and lymph nodes, similar to those frequently observed in horses, the natural hosts. Therefore, ELR1 and eCT1 are essential in vivo for EIAV invasion and replication.


Subject(s)
Cyclin T/biosynthesis , Equine Infectious Anemia/virology , Gene Expression , Infectious Anemia Virus, Equine/growth & development , Receptors, Virus/biosynthesis , Animal Structures/virology , Animals , Blotting, Western , Cyclin T/genetics , Disease Models, Animal , Equine Infectious Anemia/pathology , Gene Expression Profiling , Horses , Lymph Nodes/pathology , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptors, Virus/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spleen/pathology , Virus Replication
3.
AIDS Res Hum Retroviruses ; 31(1): 137-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24528253

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase 9/metabolism , Disulfiram/pharmacology , Hydroxamic Acids/pharmacology , Positive Transcriptional Elongation Factor B/metabolism , Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Cell Line , Cyclin T/biosynthesis , HIV Infections/virology , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Jurkat Cells , Phosphorylation/drug effects , Virus Activation/drug effects , Virus Latency , Vorinostat
4.
Immunity ; 41(2): 325-38, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25148027

ABSTRACT

Classical genetic approaches to examine the requirements of genes for T cell differentiation during infection are time consuming. Here we developed a pooled approach to screen 30-100+ genes individually in separate antigen-specific T cells during infection using short hairpin RNAs in a microRNA context (shRNAmir). Independent screens using T cell receptor (TCR)-transgenic CD4(+) and CD8(+) T cells responding to lymphocytic choriomeningitis virus (LCMV) identified multiple genes that regulated development of follicular helper (Tfh) and T helper 1 (Th1) cells, and short-lived effector and memory precursor cytotoxic T lymphocytes (CTLs). Both screens revealed roles for the positive transcription elongation factor (P-TEFb) component Cyclin T1 (Ccnt1). Inhibiting expression of Cyclin T1, or its catalytic partner Cdk9, impaired development of Th1 cells and protective short-lived effector CTL and enhanced Tfh cell and memory precursor CTL formation in vivo. This pooled shRNA screening approach should have utility in numerous immunological studies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Lymphocytic choriomeningitis virus/immunology , RNA Interference/immunology , Animals , Cell Differentiation/genetics , Cyclin T/biosynthesis , Cyclin T/genetics , Cyclin-Dependent Kinase 9/biosynthesis , Cyclin-Dependent Kinase 9/genetics , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Positive Regulatory Domain I-Binding Factor 1 , RNA, Small Interfering , Receptors, Antigen, T-Cell/genetics , T-Box Domain Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Transcription Factors/genetics , Transduction, Genetic/methods
5.
Antimicrob Agents Chemother ; 58(11): 6558-71, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25155598

ABSTRACT

HIV-1 transcription is activated by the Tat protein, which recruits CDK9/cyclin T1 to the HIV-1 promoter. CDK9 is phosphorylated by CDK2, which facilitates formation of the high-molecular-weight positive transcription elongation factor b (P-TEFb) complex. We previously showed that chelation of intracellular iron inhibits CDK2 and CDK9 activities and suppresses HIV-1 transcription, but the mechanism of the inhibition was not understood. In the present study, we tested a set of novel iron chelators for the ability to inhibit HIV-1 transcription and elucidated their mechanism of action. Novel phenyl-1-pyridin-2yl-ethanone (PPY)-based iron chelators were synthesized and examined for their effects on cellular iron, HIV-1 inhibition, and cytotoxicity. Activities of CDK2 and CDK9, expression of CDK9-dependent and CDK2-inhibitory mRNAs, NF-κB expression, and HIV-1- and NF-κB-dependent transcription were determined. PPY-based iron chelators significantly inhibited HIV-1, with minimal cytotoxicity, in cultured and primary cells chronically or acutely infected with HIV-1 subtype B, but they had less of an effect on HIV-1 subtype C. Iron chelators upregulated the expression of IκB-α, with increased accumulation of cytoplasmic NF-κB. The iron chelators inhibited CDK2 activity and reduced the amount of CDK9/cyclin T1 in the large P-TEFb complex. Iron chelators reduced HIV-1 Gag and Env mRNA synthesis but had no effect on HIV-1 reverse transcription. In addition, iron chelators moderately inhibited basal HIV-1 transcription, equally affecting HIV-1 and Sp1- or NF-κB-driven transcription. By virtue of their involvement in targeting several key steps in HIV-1 transcription, these novel iron chelators have the potential for the development of new therapeutics for the treatment of HIV-1 infection.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 9/metabolism , HIV-1/genetics , I-kappa B Kinase/biosynthesis , Iron Chelating Agents/pharmacology , Cell Line , Cell Survival , Cyclin A/biosynthesis , Cyclin A/genetics , Cyclin E/biosynthesis , Cyclin E/genetics , Cyclin T/biosynthesis , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Viral/drug effects , HEK293 Cells , HIV-1/drug effects , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , RNA, Messenger/biosynthesis , Reverse Transcription/drug effects , S Phase Cell Cycle Checkpoints/drug effects , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects , Virus Replication/drug effects , env Gene Products, Human Immunodeficiency Virus/biosynthesis , gag Gene Products, Human Immunodeficiency Virus/biosynthesis
6.
Gene ; 505(1): 1-8, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22692005

ABSTRACT

Cyclin T1 (CCNT1), a gene containing nine exons, forms the positive transcription elongation factor b (P-TEFb) complex and regulates a wide variety of biological processes including transcription. We discovered a novel splice variant of CCNT1 that lacks exon 7 (dE7). RT-PCR analysis revealed that the dE7 transcript was detected in almost all tissues examined. The dE7/FL transcript ratio was high in quiescent peripheral blood mononuclear cells (PBMC) and in tissues poor in cell division; however, it was low in activated PBMC and in tissues with high cell proliferative potential. These results suggest that exon 7 skipping is linked to cell cycle progression. Increasing the dE7/FL transcript ratio resulted in the reduction of CCNT1 protein levels, indicating that the expression of CCNT1 protein is controlled by exon skipping. Exon 7 skipping yields a +1 frameshift at exon 8, which generates a premature termination codon (PTC). The dE7 transcript levels increased when cells were treated with the protein synthesis inhibitor cycloheximide (CHX) or a kinase inhibitor wortmannin (WORT), whilst the FL transcript levels were unchanged, suggesting that the dE7 transcript is a target of nonsense-mediated decay (NMD). Importantly, reduction of dE7 transcript by WORT correlated well with the decrement of CCNT1 protein expression. The dE7 transcript would produce an approximately 23kDa protein that covers approximately 70% of the cyclin box. The ectopically expressed dE7 protein physically interacted with CDK9 and competed with FL CCNT1 for CDK9, thus should act dominant-negatively on FL CCNT1. The replication of human immunodeficiency virus type 1 (HIV-1), heavily dependent on the CCNT1 function, was inhibited by dE7 protein through the attenuation of Tat/long terminal repeat (LTR)-driven transcription. Taken together, these results suggest that dE7 is a novel splice variant that regulates the expression and function of CCNT1.


Subject(s)
Alternative Splicing/physiology , Codon, Terminator/metabolism , Cyclin T/biosynthesis , Exons/physiology , Leukocytes, Mononuclear/metabolism , RNA, Messenger/biosynthesis , Alternative Splicing/drug effects , Androstadienes/pharmacology , Cell Line , Codon, Terminator/genetics , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cycloheximide/pharmacology , Female , HIV Long Terminal Repeat/genetics , HIV-1/genetics , HIV-1/metabolism , Humans , Leukocytes, Mononuclear/cytology , Male , Protein Kinase Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Wortmannin , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
7.
Mol Endocrinol ; 24(12): 2267-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980437

ABSTRACT

Previously we determined that S81 is the highest stoichiometric phosphorylation on the androgen receptor (AR) in response to hormone. To explore the role of this phosphorylation on growth, we stably expressed wild-type and S81A mutant AR in LHS and LAPC4 cells. The cells with increased wild-type AR expression grow faster compared with parental cells and S81A mutant-expressing cells, indicating that loss of S81 phosphorylation limits cell growth. To explore how S81 regulates cell growth, we tested whether S81 phosphorylation regulates AR transcriptional activity. LHS cells stably expressing wild-type and S81A mutant AR showed differences in the regulation of endogenous AR target genes, suggesting that S81 phosphorylation regulates promoter selectivity. We next sought to identify the S81 kinase using ion trap mass spectrometry to analyze AR-associated proteins in immunoprecipitates from cells. We observed cyclin-dependent kinase (CDK)9 association with the AR. CDK9 phosphorylates the AR on S81 in vitro. Phosphorylation is specific to S81 because CDK9 did not phosphorylate the AR on other serine phosphorylation sites. Overexpression of CDK9 with its cognate cyclin, Cyclin T, increased S81 phosphorylation levels in cells. Small interfering RNA knockdown of CDK9 protein levels decreased hormone-induced S81 phosphorylation. Additionally, treatment of LNCaP cells with the CDK9 inhibitors, 5,6-dichloro-1-ß-D-ribofuranosylbenzimidazole and Flavopiridol, reduced S81 phosphorylation further, suggesting that CDK9 regulates S81 phosphorylation. Pharmacological inhibition of CDK9 also resulted in decreased AR transcription in LNCaP cells. Collectively these results suggest that CDK9 phosphorylation of AR S81 is an important step in regulating AR transcriptional activity and prostate cancer cell growth.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Receptors, Androgen/metabolism , Serine/metabolism , Androgen Receptor Antagonists/metabolism , Androgens/metabolism , Animals , COS Cells , Cell Growth Processes/genetics , Cells, Cultured , Chlorocebus aethiops , Cyclin T/biosynthesis , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/deficiency , Cyclin-Dependent Kinase 9/genetics , Dichlororibofuranosylbenzimidazole/pharmacology , Flavonoids/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , Phosphorylation , Piperidines/pharmacology , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Serine/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...