Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Mol Cell ; 83(3): 393-403, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36599353

ABSTRACT

The positive transcription elongation factor b (P-TEFb) is composed of cyclins T1 or T2 and cyclin-dependent kinase 9 that regulate the elongation phase of transcription by RNA polymerase II. By antagonizing negative elongation factors and phosphorylating the C-terminal domain of RNA polymerase II, P-TEFb facilitates the elongation and co-transcriptional processing of nascent transcripts. This step is critical for the expression of most eukaryotic genes. In growing cells, P-TEFb is regulated negatively by its reversible associations with HEXIM1/2 in the 7SK snRNP and positively by a number of transcription factors, as well as the super elongation complex. In resting cells, P-TEFb falls apart, and cyclin T1 is degraded by the proteasome. This complex regulation of P-TEFb has evolved for the precise temporal and spatial regulation of gene expression in the organism. Its dysregulation contributes to inflammatory and neoplastic conditions.


Subject(s)
Positive Transcriptional Elongation Factor B , RNA Polymerase II , Humans , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , HeLa Cells , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin T/genetics , Cyclin T/metabolism , Transcription, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Hum Mol Genet ; 33(8): 733-738, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38215789

ABSTRACT

OBJECTIVE: This study aims to identify BMI-associated genes by integrating aggregated summary information from different omics data. METHODS: We conducted a meta-analysis to leverage information from a genome-wide association study (n = 339 224), a transcriptome-wide association study (n = 5619), and an epigenome-wide association study (n = 3743). We prioritized the significant genes with a machine learning-based method, netWAS, which borrows information from adipose tissue-specific interaction networks. We also used the brain-specific network in netWAS to investigate genes potentially involved in brain-adipose interaction. RESULTS: We identified 195 genes that were significantly associated with BMI through meta-analysis. The netWAS analysis narrowed down the list to 21 genes in adipose tissue. Among these 21 genes, six genes, including FUS, STX4, CCNT2, FUBP1, NDUFS3, and RAPSN, were not reported to be BMI-associated in PubMed or GWAS Catalog. We also identified 11 genes that were significantly associated with BMI in both adipose and whole brain tissues. CONCLUSION: This study integrated three types of omics data and identified a group of genes that have not previously been reported to be associated with BMI. This strategy could provide new insights for future studies to identify molecular mechanisms contributing to BMI regulation.


Subject(s)
Genome-Wide Association Study , Multiomics , Humans , Body Mass Index , Genome-Wide Association Study/methods , Transcriptome , Obesity/genetics , Cyclin T/genetics , DNA-Binding Proteins/genetics , RNA-Binding Proteins/genetics
3.
Mol Cell ; 65(6): 1068-1080.e5, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28262505

ABSTRACT

The BET proteins are major transcriptional regulators and have emerged as new drug targets, but their functional distinction has remained elusive. In this study, we report that the BET family members Brd2 and Brd4 exert distinct genomic functions at genes whose transcription they co-regulate during mouse T helper 17 (Th17) cell differentiation. Brd2 is associated with the chromatin insulator CTCF and the cohesin complex to support cis-regulatory enhancer assembly for gene transcriptional activation. In this context, Brd2 binds the transcription factor Stat3 in an acetylation-sensitive manner and facilitates Stat3 recruitment to active enhancers occupied with transcription factors Irf4 and Batf. In parallel, Brd4 temporally controls RNA polymerase II (Pol II) processivity during transcription elongation through cyclin T1 and Cdk9 recruitment and Pol II Ser2 phosphorylation. Collectively, our study uncovers both separate and interdependent Brd2 and Brd4 functions in potentiating the genetic program required for Th17 cell development and adaptive immunity.


Subject(s)
Adaptive Immunity , Cell Differentiation , Chromatin/enzymology , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Th17 Cells/enzymology , Transcription Factors/metabolism , Transcription, Genetic , Acetylation , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin/genetics , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice, Inbred C57BL , Models, Molecular , Nuclear Proteins/genetics , Phenotype , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , RNA Polymerase II/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Structure-Activity Relationship , Th17 Cells/immunology , Transcription Factors/genetics , Transfection , Cohesins
4.
Nucleic Acids Res ; 50(9): 5000-5013, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35524561

ABSTRACT

P-TEFb, composed of CycT1 and CDK9, regulates the elongation of transcription by RNA polymerase II. In proliferating cells, it is regulated by 7SK snRNA in the 7SK snRNP complex. In resting cells, P-TEFb is absent, because CycT1 is dephosphorylated, released from CDK9 and rapidly degraded. In this study, we identified the mechanism of this degradation. We mapped the ubiquitination and degradation of free CycT1 to its N-terminal region from positions 1 to 280. This region is ubiquitinated at six lysines, where E3 ligases Siah1 and Siah2 bind and degrade these sequences. Importantly, the inhibition of Siah1/2 rescued the expression of free CycT1 in proliferating as well as resting primary cells. We conclude that Siah1/2 are the E3 ligases that bind and degrade the dissociated CycT1 in resting, terminally differentiated, anergic and/or exhausted cells.


Subject(s)
Positive Transcriptional Elongation Factor B , Transcription Factors , Ubiquitin-Protein Ligases/metabolism , Cell Physiological Phenomena , Cyclin T/genetics , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/metabolism , RNA, Small Nuclear , Transcription Factors/metabolism , Transcription, Genetic
5.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619107

ABSTRACT

Reactivation of human cytomegalovirus (HCMV) from latency is a major health consideration for recipients of stem-cell and solid organ transplantations. With over 200,000 transplants taking place globally per annum, virus reactivation can occur in more than 50% of cases leading to loss of grafts as well as serious morbidity and even mortality. Here, we present the most extensive screening to date of epigenetic inhibitors on HCMV latently infected cells and find that histone deacetylase inhibitors (HDACis) and bromodomain inhibitors are broadly effective at inducing virus immediate early gene expression. However, while HDACis, such as myeloid-selective CHR-4487, lead to production of infectious virions, inhibitors of bromodomain (BRD) and extraterminal proteins (I-BETs), including GSK726, restrict full reactivation. Mechanistically, we show that BET proteins (BRDs) are pivotally connected to regulation of HCMV latency and reactivation. Through BRD4 interaction, the transcriptional activator complex P-TEFb (CDK9/CycT1) is sequestered by repressive complexes during HCMV latency. Consequently, I-BETs allow release of P-TEFb and subsequent recruitment to promoters via the superelongation complex (SEC), inducing transcription of HCMV lytic genes encoding immunogenic antigens from otherwise latently infected cells. Surprisingly, this occurs without inducing many viral immunoevasins and, importantly, while also restricting viral DNA replication and full HCMV reactivation. Therefore, this pattern of HCMV transcriptional dysregulation allows effective cytotoxic immune targeting and killing of latently infected cells, thus reducing the latent virus genome load. This approach could be safely used to pre-emptively purge the virus latent reservoir prior to transplantation, thereby reducing HCMV reactivation-related morbidity and mortality.


Subject(s)
Cell Cycle Proteins/genetics , Cytomegalovirus/immunology , DNA, Viral/genetics , Epigenesis, Genetic , Histone Deacetylases/genetics , Positive Transcriptional Elongation Factor B/genetics , Transcription Factors/genetics , Azepines/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzodiazepines/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/immunology , Cyclin T/genetics , Cyclin T/immunology , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/immunology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , DNA Replication/drug effects , DNA, Viral/antagonists & inhibitors , DNA, Viral/immunology , Genes, Immediate-Early , Genes, Reporter , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/immunology , Host-Pathogen Interactions , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Models, Biological , Positive Transcriptional Elongation Factor B/immunology , Primary Cell Culture , Promoter Regions, Genetic , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , THP-1 Cells , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/immunology , Transcription, Genetic , Virus Activation/drug effects , Virus Latency/drug effects
6.
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
7.
Proc Natl Acad Sci U S A ; 117(27): 15763-15771, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571938

ABSTRACT

HIV-1 latency is a major barrier to cure. Identification of small molecules that destabilize latency and allow immune clearance of infected cells could lead to treatment-free remission. In vitro models of HIV-1 latency involving cell lines or primary cells have been developed for characterization of HIV-1 latency and high-throughput screening for latency-reversing agents (LRAs). We have shown that the majority of LRAs identified to date are relatively ineffective in cells from infected individuals despite activity in model systems. We show here that, for diverse LRAs, latency reversal observed in model systems involves a heat shock factor 1 (HSF1)-mediated stress pathway. Small-molecule inhibition of HSF1 attenuated HIV-1 latency reversal by histone deactylase inhibitors, protein kinase C agonists, and proteasome inhibitors without interfering with the known mechanism of action of these LRAs. However, latency reversal by second mitochondria-derived activator of caspase (SMAC) mimetics was not affected by inhibition of HSF1. In cells from infected individuals, inhibition of HSF1 attenuated latency reversal by phorbol ester+ionomycin but not by anti-CD3+anti-CD28. HSF1 promotes elongation of HIV-1 RNA by recruiting P-TEFb to the HIV-1 long terminal repeat (LTR), and we show that inhibition of HSF1 attenuates the formation of elongated HIV-1 transcripts. We demonstrate that in vitro models of latency have higher levels of the P-TEFb subunit cyclin T1 than primary cells, which may explain why many LRAs are functional in model systems but relatively ineffective in primary cells. Together, these studies provide insights into why particular LRA combinations are effective in reversing latency in cells from infected individuals.


Subject(s)
HIV Infections/genetics , HIV-1/genetics , Heat Shock Transcription Factors/genetics , Virus Latency/genetics , Anti-HIV Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Cyclin T/genetics , HIV Infections/virology , HIV-1/pathogenicity , Heat Shock Transcription Factors/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Humans , Mitochondrial Proteins/genetics , Positive Transcriptional Elongation Factor B/genetics , Protein Kinase C/genetics , RNA, Viral/drug effects , RNA, Viral/genetics , Small Molecule Libraries/pharmacology , Terminal Repeat Sequences/genetics , Virus Activation/genetics
8.
J Virol ; 95(13): e0046021, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33827943

ABSTRACT

Ubiquitination plays an important role in human immunodeficiency virus 1 (HIV-1) infection. HIV proteins such as Vif and Vpx mediate the degradation of the host proteins APOBEC3 and SAMHD1, respectively, through the proteasome pathway. However, whether deubiquitylating enzymes play an essential role in HIV-1 infection is largely unknown. Here, we demonstrate that the deubiquitinase USP21 potently inhibits HIV-1 production by indirectly downregulating the expression of HIV-1 transactivator of transcription (Tat), which is essential for transcriptional elongation in HIV-1. USP21 deubiquitylates Tat via its deubiquitinase activity, but a stronger ability to reduce Tat expression than a dominant-negative ubiquitin mutant (Ub-KO) showed that other mechanisms may contribute to USP21-mediated inhibition of Tat. Further investigation showed that USP21 downregulates cyclin T1 mRNA levels by increasing methylation of histone K9 in the promoter of cyclin T1, a subunit of the positive transcription elongation factor b (P-TEFb) that interacts with Tat and transactivation response element (TAR) and is required for transcription stimulation and Tat stability. Moreover, USP21 had no effect on the function of other HIV-1 accessory proteins, including Vif, Vpr, Vpx, and Vpu, indicating that USP21 was specific to Tat. These findings improve our understanding of USP21-mediated functional suppression of HIV-1 production. IMPORTANCE Ubiquitination plays an essential role in viral infection. Deubiquitinating enzymes (DUBs) reverse ubiquitination by cleaving ubiquitins from target proteins, thereby affecting viral infection. The role of the members of the USP family, which comprises the largest subfamily of DUBs, is largely unknown in HIV-1 infection. Here, we screened a series of USP members and found that USP21 inhibits HIV-1 production by specifically targeting Tat but not the other HIV-1 accessory proteins. Further investigations revealed that USP21 reduces Tat expression in two ways. First, USP21 deubiquitinates polyubiquitinated Tat, causing Tat instability, and second, USP21 reduces the mRNA levels of cyclin T1 (CycT1), an important component of P-TEFb, that leads to Tat downregulation. Thus, in this study, we report a novel role of the deubiquitinase, USP21, in HIV-1 infection. USP21 represents a potentially useful target for the development of novel anti-HIV drugs.


Subject(s)
Cyclin T/metabolism , Deubiquitinating Enzymes/metabolism , HIV-1/growth & development , Ubiquitin Thiolesterase/metabolism , tat Gene Products, Human Immunodeficiency Virus/biosynthesis , Cyclin T/genetics , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Jurkat Cells , Promoter Regions, Genetic/genetics , Protein Biosynthesis/genetics , RNA, Messenger/analysis , Virus Replication/genetics
9.
Acta Pharmacol Sin ; 43(7): 1633-1645, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34811514

ABSTRACT

Cyclin-dependent kinase (CDK) 9 associates mainly with cyclin T1 and forms the positive transcription elongation factor b (p-TEFb) complex responsible for transcriptional regulation. It has been shown that CDK9 modulates the expression and activity of oncogenes, such as MYC and murine double minute 4 (MDM4), and it also plays an important role in development and/or maintenance of the malignant cell phenotype. Malfunction of CDK9 is frequently observed in numerous cancers. Recent studies have highlighted the function of CDK9 through a variety of mechanisms in cancers, including the formation of new complexes and epigenetic alterations. Due to the importance of CDK9 activation in cancer cells, CDK9 inhibitors have emerged as promising candidates for cancer therapy. Natural product-derived and chemically synthesized CDK9 inhibitors are being examined in preclinical and clinical research. In this review, we summarize the current knowledge on the role of CDK9 in transcriptional regulation, epigenetic regulation, and different cellular factor interactions, focusing on new advances. We show the importance of CDK9 in mediating tumorigenesis and tumor progression. Then, we provide an overview of some CDK9 inhibitors supported by multiple oncologic preclinical and clinical investigations. Finally, we discuss the perspective and challenge of CDK9 modulation in cancer.


Subject(s)
Cyclin-Dependent Kinase 9 , Neoplasms , Animals , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Mice , Neoplasms/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Transcription, Genetic
10.
Int J Mol Sci ; 23(19)2022 Oct 05.
Article in English | MEDLINE | ID: mdl-36233116

ABSTRACT

The complex host interaction network of human cytomegalovirus (HCMV) involves the regulatory protein kinase pUL97, which represents a viral cyclin-dependent kinase (CDK) ortholog. pUL97 interacts with the three human cyclin types T1, H, and B1, whereby the binding region of cyclin T1 and the pUL97 oligomerization region were both assigned to amino acids 231-280. We further addressed the question of whether HCMVs harboring mutations in ORF-UL97, i.e., short deletions or resistance-conferring point mutations, are affected in the interaction with human cyclins and viral replication. To this end, clinically relevant UL97 drug-resistance-conferring mutants were analyzed by whole-genome sequencing and used for genetic marker transfer experiments. The recombinant HCMVs indicated conservation of pUL97-cyclin interaction, since all viral UL97 point mutants continued to interact with the analyzed cyclin types and exerted wild-type-like replication fitness. In comparison, recombinant HCMVs UL97 Δ231-280 and also the smaller deletion Δ236-275, but not Δ241-270, lost interaction with cyclins T1 and H, showed impaired replication efficiency, and also exhibited reduced kinase activity. Moreover, a cellular knock-out of cyclins B1 or T1 did not alter HCMV replication phenotypes or pUL97 kinase activity, possibly indicating alternative, compensatory pUL97-cyclin interactions. In contrast, however, cyclin H knock-out, similar to virus deletion mutants in the pUL97-cyclin H binding region, exhibited strong defective phenotypes of HCMV replication, as supported by reduced pUL97 kinase activity in a cyclin H-dependent coexpression setting. Thus, cyclin H proved to be a very relevant determinant of pUL97 kinase activity and viral replication efficiency. As a conclusion, the results provide evidence for the functional importance of pUL97-cyclin interaction. High selective pressure on the formation of pUL97-cyclin complexes was identified by the use of clinically relevant mutants.


Subject(s)
Cyclin H , Cytomegalovirus , Viral Proteins , Amino Acids/metabolism , Cyclin H/genetics , Cyclin H/metabolism , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytomegalovirus/physiology , Genetic Markers , Humans , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Viral Proteins/genetics , Virus Replication/genetics
11.
Nucleic Acids Res ; 47(9): 4462-4475, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30864669

ABSTRACT

The general transcription factor P-TEFb, a master regulator of RNA polymerase (Pol) II elongation, phosphorylates the C-terminal domain (CTD) of Pol II and negative elongation factors to release Pol II from promoter-proximal pausing. We show here that P-TEFb surprisingly inhibits the myoblast differentiation into myotubes, and that P-TEFb and its two positive complexes are eliminated in this process. In contrast, DYRK1A, another CTD kinase known to control transcription of a subset of genes important for development and tissue homeostasis, is found to activate transcription of key myogenic genes. We show that active DYRK1A exists in a complex with the WD40-repeat protein DCAF7 that stabilizes and tethers DYRK1A to Pol II, so that DYRK1A-DCAF7 can co-migrate with and phosphorylate Pol II along the myogenic gene loci. Thus, DCAF7 modulates the kinase signaling output of DYRK1A on Pol II to stimulate myogenic transcription after active P-TEFb function is shut off.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Muscle Development/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Transcription, Genetic , Animals , Cell Differentiation/genetics , Cyclin T/genetics , Cyclin-Dependent Kinase 9/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Nuclear Proteins/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , SEC Translocation Channels/genetics , Transcription Factors/genetics , Dyrk Kinases
12.
J Biol Chem ; 294(15): 6188-6203, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30782840

ABSTRACT

Human cytomegalovirus (HCMV) is a common ß-herpesvirus causing life-long latent infections. HCMV replication interferes with cell cycle regulation in host cells because the HCMV-encoded cyclin-dependent kinase (CDK) ortholog pUL97 extensively phosphorylates the checkpoint regulator retinoblastoma protein. pUL97 also interacts with cyclins B1, T1, and H, and recent findings have strongly suggested that these interactions influence pUL97 substrate recognition. Interestingly, here we detected profound mechanistic differences among these pUL97-cyclin interactions. Our study revealed the following. (i) pUL97 interacts with cyclins B1 and H in a manner dependent on pUL97 activity and HCMV-specific cyclin modulation, respectively. (ii) The phosphorylated state of both proteins is an important determinant of the pUL97-cyclin B1 interaction. (iii) Activated phospho-Thr-315 cyclin H is up-regulated during HCMV replication. (iv) Thr-315 phosphorylation is independent of intracellular pUL97 or CDK7 activity. (v) pUL97-mediated in vitro phosphorylation is detectable for cyclin B1 but not H. (vi) Mutual transphosphorylation between pUL97 and CDK7 is not detectable, and an MS-based phosphosite analysis indicated that pUL97 might unexpectedly not be phosphorylated in its T-loop. (vii) The binary complexes pUL97-cyclin H and CDK7-cyclin H as well as the ternary complex pUL97-cyclin-H-CDK7 are detectable in an assembly-based CoIP approach. (viii) pUL97 self-interaction can be bridged by the transcriptional cyclins T1 or H but not by the classical cell cycle-regulating B1 cyclin. Combined, our findings unravel a number of cyclin type-specific differences in pUL97 interactions and suggest a multifaceted regulatory impact of cyclins on HCMV replication.


Subject(s)
Cyclin B1/metabolism , Cyclin H/metabolism , Cyclin T/metabolism , Cytomegalovirus/physiology , Viral Proteins/metabolism , Virus Replication/physiology , Cyclin B1/genetics , Cyclin H/genetics , Cyclin T/genetics , HEK293 Cells , Humans , Phosphorylation , Protein Domains , Protein Structure, Quaternary , Viral Proteins/genetics
13.
Anticancer Drugs ; 31(6): 623-631, 2020 07.
Article in English | MEDLINE | ID: mdl-32058347

ABSTRACT

Gastric cancer has become the second most common malignant tumor in the world, revealing the molecular mechanism of gastric cancer development is essential for the treatment of gastric cancer and improvement of prognosis. Recent studies have shown that microRNAs may play a carcinogenic or tumor-suppressive role in many types of cancer. It has been detected that miR-216b is down-regulated in many cancer types, indicating that miR-216b can be used as a prognostic marker for these particular types of cancer. However, the effect of miR-216b on gastric cancer remains unclear. In the present study, miR-216 was observed to be significantly down-regulated in cancer tissues compared to normal tissues, and the level of miR-216b in various gastric cancer cell lines was decreased. In addition, miR-216b overexpression inhibits proliferation, migration, invasion, cell cycle and apoptosis of gastric cancer cells. We further verified that the inhibitory effect of miR-216b on proliferation and invasion of gastric cancer cells is mediated by cyclin T2. Overexpression of cyclin T2 can reverse the anti-cancer effect of miR-216b mimics. The results further enriched the mechanism of miR-216b in the development and progression of gastric cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle , Cell Proliferation , Cyclin T/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Stomach Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cyclin T/genetics , Humans , Neoplasm Invasiveness , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
14.
J Biol Chem ; 293(44): 17008-17020, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30209133

ABSTRACT

Vascular calcification (or mineralization) is a common complication of chronic kidney disease (CKD) and is closely associated with increased mortality and morbidity rates. We recently reported that activation of the activating transcription factor 4 (ATF4) pathway through the saturated fatty acid (SFA)-induced endoplasmic reticulum (ER) stress response plays a causative role in CKD-associated vascular calcification. Here, using mouse models of CKD, we 1) studied the contribution of the proapoptotic transcription factor CCAAT enhancer-binding protein homologous protein (CHOP) to CKD-dependent medial calcification, and 2) we identified an additional regulator of ER stress-mediated CHOP expression. Transgenic mice having smooth muscle cell (SMC)-specific CHOP expression developed severe vascular apoptosis and medial calcification under CKD. Screening of a protein kinase inhibitor library identified 16 compounds, including seven cyclin-dependent kinase (CDK) inhibitors, that significantly suppressed CHOP induction during ER stress. Moreover, selective CDK9 inhibitors and CRISPR/Cas9-mediated CDK9 reduction blocked SFA-mediated induction of CHOP expression, whereas inhibitors of other CDK isoforms did not. Cyclin T1 knockout inhibited SFA-mediated induction of CHOP and mineralization, whereas deletion of cyclin T2 and cyclin K promoted CHOP expression levels and mineralization. Of note, the CDK9-cyclin T1 complex directly phosphorylated and activated ATF4. These results demonstrate that the CDK9-cyclin T1 and CDK9-cyclin T2/K complexes have opposing roles in CHOP expression and CKD-induced vascular calcification. They further reveal that the CDK9-cyclin T1 complex mediates vascular calcification through CHOP induction and phosphorylation-mediated ATF4 activation.


Subject(s)
Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , Fatty Acids/metabolism , Renal Insufficiency, Chronic/complications , Transcription Factor CHOP/genetics , Vascular Calcification/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Cyclin T/genetics , Cyclin-Dependent Kinase 9/genetics , Endoplasmic Reticulum Stress , Humans , Male , Mice , Mice, Inbred DBA , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Protein Binding , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Transcription Factor CHOP/metabolism , Vascular Calcification/etiology , Vascular Calcification/genetics , Vascular Calcification/physiopathology
15.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29321323

ABSTRACT

Cells derived from mice and other rodents exhibit profound blocks to HIV-1 virion production, reflecting species-specific incompatibilities between viral Tat and Rev proteins and essential host factors cyclin T1 (CCNT1) and exportin-1 (XPO1, also known as CRM1), respectively. To determine if mouse cell blocks other than CCNT1 and XPO1 affect HIV's postintegration stages, we studied HIV-1NL4-3 gene expression in mouse NIH 3T3 cells modified to constitutively express HIV-1-compatible versions of CCNT1 and XPO1 (3T3.CX cells). 3T3.CX cells supported both Rev-independent and Rev-dependent viral gene expression and produced relatively robust levels of virus particles, confirming that CCNT1 and XPO1 represent the predominant blocks to these stages. Unexpectedly, however, 3T3.CX cells were remarkably resistant to virus-induced cytopathic effects observed in human cell lines, which we mapped to the viral protein Vif and its apparent species-specific capacity to induce G2/M cell cycle arrest. Vif was able to mediate rapid degradation of human APOBEC3G and the PPP2R5D regulatory B56 subunit of the PP2A phosphatase holoenzyme in mouse cells, thus demonstrating that VifNL4-3's modulation of the cell cycle can be functionally uncoupled from some of its other defined roles in CUL5-dependent protein degradation. Vif was also unable to induce G2/M cell cycle arrest in other nonhuman cell types, including cells derived from nonhuman primates, leading us to propose that one or more human-specific cofactors underpin Vif's ability to modulate the cell cycle.IMPORTANCE Cells derived from mice and other rodents exhibit profound blocks to HIV-1 replication, thus hindering the development of a low-cost small-animal model for studying HIV/AIDS. Here, we engineered otherwise-nonpermissive mouse cells to express HIV-1-compatible versions of two species-specific host dependency factors, cyclin T1 (CCNT1) and exportin-1 (XPO1) (3T3.CX cells). We show that 3T3.CX cells rescue HIV-1 particle production but, unexpectedly, are completely resistant to virus-induced cytopathic effects. We mapped these effects to the viral accessory protein Vif, which induces a prolonged G2/M cell cycle arrest followed by apoptosis in human cells. Combined, our results indicate that one or more additional human-specific cofactors govern HIV-1's capacity to modulate the cell cycle, with potential relevance to viral pathogenesis in people and existing animal models.


Subject(s)
G2 Phase Cell Cycle Checkpoints , HIV-1/metabolism , M Phase Cell Cycle Checkpoints , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase/genetics , APOBEC-3G Deaminase/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Cyclin T/genetics , Cyclin T/metabolism , HIV-1/genetics , HeLa Cells , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice , NIH 3T3 Cells , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Species Specificity , vif Gene Products, Human Immunodeficiency Virus/genetics , Exportin 1 Protein
16.
Virol J ; 16(1): 22, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30786885

ABSTRACT

BACKGROUND: The regulatory cyclin, Cyclin T1 (CycT1), is a host factor essential for HIV-1 replication in CD4 T cells and macrophages. The importance of CycT1 and the Positive Transcription Elongation Factor b (P-TEFb) complex for HIV replication is well-established, but regulation of CycT1 expression and protein levels during HIV replication and latency establishment in CD4 T cells is less characterized. METHODS: To better define the regulation of CycT1 levels during HIV replication in CD4 T cells, multiparameter flow cytometry was utilized to study the interaction between HIV replication (intracellular p24) and CycT1 of human peripheral blood memory CD4 T cells infected with HIV in vitro. CycT1 was further examined in CD4 T cells of human lymph nodes. RESULTS: In activated (CD3+CD28 costimulation) uninfected blood memory CD4 T cells, CycT1 was most significantly upregulated in maximally activated (CD69+CD25+ and HLA.DR+CD38+) cells. In memory CD4 T cells infected with HIV in vitro, two distinct infected populations of p24+CycT1+ and p24+CycT1- cells were observed during 7 days infection, suggestive of different phases of productive HIV replication and subsequent latency establishment. Intriguingly, p24+CycT1- cells were the predominant infected population in activated CD4 T cells, raising the possibility that productively infected cells may transition into latency subsequent to CycT1 downregulation. Additionally, when comparing infected p24+ cells to bystander uninfected p24- cells (after bulk HIV infections), HIV replication significantly increased T cell activation (CD69, CD25, HLA.DR, CD38, and Ki67) without concomitantly increasing CycT1 protein levels, possibly due to hijacking of P-TEFb by the viral Tat protein. Lastly, CycT1 was constitutively expressed at higher levels in lymph node CD4 T cells compared to blood T cells, potentially enhancing latency generation in lymphoid tissues. CONCLUSIONS: CycT1 is most highly upregulated in maximally activated memory CD4 T cells as expected, but may become less associated with T cell activation during HIV replication. The progression into latency may further be predicated by substantial generation of p24+CycT1- cells during HIV replication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cyclin T/genetics , HIV Infections/immunology , Virus Latency/physiology , Virus Replication/physiology , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Gene Expression Regulation , HIV-1/physiology , Host Microbial Interactions , Humans , Positive Transcriptional Elongation Factor B/genetics , Transcriptional Activation
17.
J Cell Biochem ; 119(2): 1273-1284, 2018 02.
Article in English | MEDLINE | ID: mdl-28722178

ABSTRACT

Cyclin-Dependent Kinase 9 (CDK9) is part of a functional diverse group of enzymes responsible for cell cycle control and progression. It associates mainly with Cyclin T1 and forms the Positive Transcription Elongation Factor b (p-TEFb) complex responsible for regulation of transcription elongation and mRNA maturation. Recent studies have highlighted the importance of CDK9 in many relevant pathologic processes, like cancer, cardiovascular diseases, and viral replication. Herein we provide an overview of the different pathways in which CDK9 is directly and indirectly involved.


Subject(s)
Cardiovascular Diseases/metabolism , Cyclin-Dependent Kinase 9/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Virus Diseases/metabolism , Animals , Cardiovascular Diseases/genetics , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Humans , Neoplasm Proteins/genetics , Neoplasms/genetics , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Transcription Elongation, Genetic , Virus Diseases/genetics
18.
Retrovirology ; 15(1): 39, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29792216

ABSTRACT

BACKGROUND: HIV-1 transcription activator protein Tat is phosphorylated in vitro by CDK2 and DNA-PK on Ser-16 residue and by PKR on Tat Ser-46 residue. Here we analyzed Tat phosphorylation in cultured cells and its functionality. RESULTS: Mass spectrometry analysis showed primarily Tat Ser-16 phosphorylation in cultured cells. In vitro, CDK2/cyclin E predominantly phosphorylated Tat Ser-16 and PKR-Tat Ser-46. Alanine mutations of either Ser-16 or Ser-46 decreased overall Tat phosphorylation. Phosphorylation of Tat Ser-16 was reduced in cultured cells treated by a small molecule inhibitor of CDK2 and, to a lesser extent, an inhibitor of DNA-PK. Conditional knock-downs of CDK2 and PKR inhibited and induced one round HIV-1 replication respectively. HIV-1 proviral transcription was inhibited by Tat alanine mutants and partially restored by S16E mutation. Pseudotyped HIV-1 with Tat S16E mutation replicated well, and HIV-1 Tat S46E-poorly, but no live viruses were obtained with Tat S16A or Tat S46A mutations. TAR RNA binding was affected by Tat Ser-16 alanine mutation. Binding to cyclin T1 showed decreased binding of all Ser-16 and Ser-46 Tat mutants with S16D and Tat S46D mutationts showing the strongest effect. Molecular modelling and molecular dynamic analysis revealed significant structural changes in Tat/CDK9/cyclin T1 complex with phosphorylated Ser-16 residue, but not with phosphorylated Ser-46 residue. CONCLUSION: Phosphorylation of Tat Ser-16 induces HIV-1 transcription, facilitates binding to TAR RNA and rearranges CDK9/cyclin T1/Tat complex. Thus, phosphorylation of Tat Ser-16 regulates HIV-1 transcription and may serve as target for HIV-1 therapeutics.


Subject(s)
Gene Expression Regulation, Viral , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Serine/metabolism , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Cells, Cultured , Cyclin T/chemistry , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Gene Knockdown Techniques , HIV Infections/genetics , Host-Pathogen Interactions , Humans , Models, Biological , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , RNA, Viral , Ubiquitination , Virus Replication , eIF-2 Kinase/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics
19.
Genes Cells ; 22(5): 424-435, 2017 May.
Article in English | MEDLINE | ID: mdl-28326644

ABSTRACT

We developed transgenic (Tg) rats that express human CD4, CCR5, CXCR4, CyclinT1, and CRM1 genes. Tg rat macrophages were efficiently infected with HIV-1 and supported production of infectious progeny virus. By contrast, both rat primary CD4+ T cells and established T cell lines expressing human CD4, CCR5, CyclinT1, and CRM1 genes were infected inefficiently, but this was ameliorated by inhibition of cyclophilin A. The infectivity of rat T cell-derived virus was lower than that of human T cell-derived virus.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclin T/metabolism , HIV Infections/immunology , Karyopherins/metabolism , Macrophages/immunology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , CD4-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Cyclin T/genetics , Disease Susceptibility , HIV-1/pathogenicity , Humans , Karyopherins/genetics , Macrophages/virology , Rats , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Exportin 1 Protein
20.
Genes Dev ; 24(20): 2303-16, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20952539

ABSTRACT

Drosophila contains one (dCDK12) and humans contain two (hCDK12 and hCDK13) proteins that are the closest evolutionary relatives of yeast Ctk1, the catalytic subunit of the major elongation-phase C-terminal repeat domain (CTD) kinase in Saccharomyces cerevisiae, CTDK-I. However, until now, neither CDK12 nor CDK13 has been demonstrated to be a bona fide CTD kinase. Using Drosophila, we demonstrate that dCDK12 (CG7597) is a transcription-associated CTD kinase, the ortholog of yCtk1. Fluorescence microscopy reveals that the distribution of dCDK12 on formaldehyde-fixed polytene chromosomes is virtually identical to that of hyperphosphorylated RNA polymerase II (RNAPII), but is distinct from that of P-TEFb (dCDK9 + dCyclin T). Chromatin immunoprecipitation (ChIP) experiments confirm that dCDK12 is present on the transcribed regions of active Drosophila genes. Compared with P-TEFb, dCDK12 amounts are lower at the 5' end and higher in the middle and at the 3' end of genes (both normalized to RNAPII). Appropriately, Drosophila dCDK12 purified from nuclear extracts manifests CTD kinase activity in vitro. Intriguingly, we find that cyclin K is associated with purified dCDK12, implicating it as the cyclin subunit of this CTD kinase. Most importantly, we demonstrate that RNAi knockdown of dCDK12 in S2 cells alters the phosphorylation state of the CTD, reducing its Ser2 phosphorylation levels. Similarly, in human HeLa cells, we show that hCDK13 purified from nuclear extracts displays CTD kinase activity in vitro, as anticipated. Also, we find that chimeric (yeast/human) versions of Ctk1 containing the kinase homology domains of hCDK12/13 (or hCDK9) are functional in yeast cells (and also in vitro); using this system, we show that a bur1(ts) mutant is rescued more efficiently by a hCDK9 chimera than by a hCDK13 chimera, suggesting the following orthology relationships: Bur1 ↔ CDK9 and Ctk1 ↔ CDK12/13. Finally, we show that siRNA knockdown of hCDK12 in HeLa cells results in alterations in the CTD phosphorylation state. Our findings demonstrate that metazoan CDK12 and CDK13 are CTD kinases, and that CDK12 is orthologous to yeast Ctk1.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Drosophila Proteins/metabolism , Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Animals , Blotting, Western , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line , Chromosome Mapping , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Genetic Complementation Test , HeLa Cells , Humans , Microscopy, Fluorescence , Mutation , Phosphorylation , Protein Kinases/genetics , RNA Interference , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL