Iron Chelation, Cell Cycle-Dependent Kinases and HIV-1 Transcription.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1550-1550
Author(s):  
Tatyana Ammosova ◽  
Zufan Debebe ◽  
Xiaomei Niu ◽  
Des R. Richardson ◽  
Marina Jerebtsova ◽  
...  

Abstract Iron chelation leads to reduced cell cycle-dependent kinase 2 (CDK2) activity (reviewed in Biochim Biophys Acta2002;1603:31–46). Elongation of HIV-1 transcription is mediated by the interaction of HIV Tat with host cell cycle-dependent kinase 9 (CDK9)/cyclin T1, which phosphorylates the C-terminal domain of RNA polymerase II, and our recent studies indicate that CDK2 is also required for Tat-dependent transcription. We hypothesized that iron chelation may inhibit HIV transcription via reduced activity of cell cycle-dependent kinases. We utilized 2-hydroxy-1-naphthylaldehyde isonicotinoyl hydrazone (311; previously shown to inhibit CDK2 expression) and 4-[3,5-bis-(hydroxyphenyl) -1,2,4-triazol-1-yl]-benzoic acid (ICL670) to chelate intracellular iron. We analyzed the effect of these chelators on HIV-1 transcription using HeLa MAGI and CEM-GFP T-cells containing an integrated HIV-1 promoter and infected with adenovirus expressing HIV-1 Tat protein. Both chelators inhibited Tat-induced HIV-1 transcription, most profoundly in CEM-GFP T-cells. The chelators also inhibited one round of HIV-1 replication in CEM-T cells infected with pseudotyped HIV-1 virus. Treatment of HeLa MAGI and CEM-GFP T-cells with iron chelators decreased CDK9 protein levels and, to a lesser extent, CDK2 protein levels. Our findings provide evidence that iron chelators may inhibit HIV-1 transcription by altering expression of CDK9 and CDK2.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3863-3863
Author(s):  
Zufan Debebe ◽  
Tatyana Ammosova ◽  
Hanspeter Nick ◽  
Xiaomei Niu ◽  
Marina Jerebtsova ◽  
...  

Abstract HIV-1 replication is induced by the excess of iron and iron chelation by desferrioxamine (DFO) inhibits viral replication in HIV-1 infected CEM T cells [1]. Treatment of cells with DFO or 2-hydroxy-1-naphthylaldehyde isonicotinoyl hydrazone inhibits expression of proteins that regulate cell-cycle progression, including cycle-dependent kinase 2 (CDK2) [2]. HIV-1 transcription is activated by Tat protein, which recruits transcriptional co-activators to the HIV-1 promoter. Elongation of HIV-1 transcription is mediated by the interaction of HIV Tat with host cell cycle-dependent kinase 9 (CDK9)/cyclin T1, which phosphorylates the C-terminal domain of RNA polymerase II. Our recent studies showed that CDK2 participates in HIV-1 transcription by phosphorylating Tat [3]. Thus inhibition of CDK2 by iron chelators might present a new approach to inhibit HIV-1 transcription. We evaluated the effect of a clinically approved orally effective iron chelator, 4-[3,5-bis-(hydroxyphenyl) -1,2,4-triazol-1-yl]-benzoic acid (ICL670 or deferasirox) on HIV-1 transcription. ICL670 inhibited Tat-induced HIV-1 transcription in CEM, 293T and HeLa cells at concentrations that did not induce cytotoxicity. The chelator decreased cellular activity of CDK2 but not its protein level and reduced HIV-1 Tat phosphorylation by CDK2. ICL670 did not decrease CDK9 protein level but significantly reduced association of CDK9 with cyclin T1 and reduced phosphorylation of Ser-2 residues of RNA polymerase II C-terminal domain. In conclusion, our findings add to the evidence that iron chelators may inhibit HIV-1 transcription by deregulating CDK2 and Cdk9. Further consideration should be given to the evaluation of ICL670 for future anti-retroviral therapeutics and to the development of iron chelators specifically as anti-retroviral agents.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 79-79 ◽  
Author(s):  
Zufan Debebe ◽  
Krishna Kumar ◽  
Tatiana Ammosova ◽  
Xiaomei Niu ◽  
Des R Richardson ◽  
...  

Abstract HIV-1 transcription is activated by HIV-1 Tat protein, which recruits transcriptional co-activators to the HIV-1 promoter. Elongation of HIV-1 transcription is mediated by the interaction of Tat with host cell cycle-dependent kinase 9 (CDK9)/cyclin T1, which phosphorylates the C-terminal domain of RNA polymerase II. Tat itself is phosphorylated by host cell cycle-dependent kinase 2 (CDK2) [1] and inhibition of CDK2 by tridentate iron chelators such as 2-hydroxy-1-naphthylaldehyde isonicotinoyl hydrazone, (311) or ICL670 (deferasirox) inhibits HIV-1 transcription [2]. In addition to the inhibition of CDK2, 311 and ICL670 also prevent association of CDK9 with cyclin T1 [2], which could lead to inhibition of CDK9 activity and also to inhibition of HIV-1 transcription. Recently, a group of novel di-2-pyridylketone thiosemicarbazone (DpT) based tridentate iron chelators were shown to exhibit marked antiproliferative activity in vivo [3]. Here we screened DpT-based and also 2-benzoylpyridine thiosemicarbazone (BpT)-based tridentate iron chelators and identified three chelators, Dp44mT, Bp4eT and Bp4aT, that inhibited HIV-1 transcription but were not cytotoxic as determined by propidium iodide uptake, LDH release and calcein-AM uptake. The inhibition of HIV-1 transcription was observed in CEM HIV-1 LTR-GFP cells infected with Adeno-Tat and in 293T cells transiently transfected with HIV-1-LTR LacZ and Tat-expressing vectors with IC50s in the mid-nanomolar range. These new iron chelators also inhibited HIV-1 replication in CEM and THP-1 cells at 10 mM concentration. Analysis of the molecular mechanism of HIV-1 inhibition revealed that the DpT- and BpT-based iron chelators inhibited the activities of both CDK9/cyclin T1 and CDK2. The CDK9/cyclin T1 complex was disrupted in the cells treated with iron chelators, suggesting a possible mechanism for the inhibition of CDK9. In conclusion, our findings provide further evidence that iron chelators may inhibit HIV-1 transcription by deregulating CDK2 and CDK9. The projected therapeutic index of the selected DpT-based iron chelators was over 103 suggesting their potential usefulness as future anti-retroviral therapeutics.


2001 ◽  
Vol 75 (23) ◽  
pp. 11336-11343 ◽  
Author(s):  
Romi Ghose ◽  
Li-Ying Liou ◽  
Christine H. Herrmann ◽  
Andrew P. Rice

ABSTRACT Combinations of cytokines are known to reactivate transcription and replication of latent human immunodeficiency virus type 1 (HIV-1) proviruses in resting CD4+ T lymphocytes isolated from infected individuals. Transcription of the HIV-1 provirus by RNA polymerase II is strongly stimulated by the viral Tat protein. Tat function is mediated by a cellular protein kinase known as TAK (cyclin T1/P-TEFb) that is composed of Cdk9 and cyclin T1. We have found that treatment of peripheral blood lymphocytes and purified resting CD4+ T lymphocytes with the combination of interleukin-2 (IL-2), IL-6, and tumor necrosis factor alpha resulted in an increase in Cdk9 and cyclin T1 protein levels and an increase in TAK enzymatic activity. The cytokine induction of TAK in resting CD4+ T lymphocytes did not appear to require proliferation of lymphocytes. These results suggest that induction of TAK by cytokines secreted in the microenvironment of lymphoid tissue may be involved in the reactivation of HIV-1 in CD4+ T lymphocytes harboring a latent provirus.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1052-1052
Author(s):  
Namita Kumari ◽  
Min Xu ◽  
Dmytro Kovlaskyy ◽  
Subhash Dhawan ◽  
Sergei Nekhai

Abstract Abstract 1052 HIV-1 transcription is activated by HIV-1 Tat protein, which recruits CDK9/cyclin T1 and other host transcriptional co-activators to the HIV-1 promoter. Tat itself is phosphorylated by cell cycle kinase 2 (CDK2) and inhibition of CDK2 by small interfering RNA or iron chelators inhibits HIV-1 transcription. HIV-1 transcription is also activated by NF-kB that binds to HIV-1 LTR independent to Tat but can also be recruited Tat-dependently by CDK9/cyclin T1. Recently, induction of heme oxygenase-1 (HO-1) by hemin was shown to inhibit HIV-1 in vitro and in vivo. Here, we analyzed the effect of novel phenyl-1-pyridin-2yl-ethanone (PPY) based iron chelators, PPYeT and PPYaT, on HIV-1. Both chelators efficiently inhibited one round of HIV-1 replication in T cells at low nanomolar IC50s without exhibiting cytotoxicity at 24 hrs incubation. The iron chelators efficiently bound intracellular labile iron as it was determined in calcein binding assays. Because we previously showed that iron chelators inhibited the activity of CDK9, we analyzed expression of several cellular genes dependent on CDK9. Unexpectedly, chelators were found to induce the expression of IkBα, an inhibitor of NF-kB (Fig1). Treatment with the iron chelators retained NF-kB in cytoplasm of the treated cells suggesting reduction in NF-kB in nucleus (Fig2). The chelators were also shown to induce HO-1 expression in cultured monocytes, likely to do a decrease of intracellular iron pool. This effect of iron chelators mimicked the effect of hemin treatment which also induced HO-1 and inhibited HIV-1 infection in our experimental conditions. Low nanomolar IC50s for the PPY-based iron chelators and lack of toxicity suggest their potential usefulness as future anti-retroviral therapeutics. Further studies are needed to investigate additional targets for iron chelators in HIV-1 life cycle that may include reverse transcription and capsid assembly. Therefore iron chelators need to be carefully assessed not only to understand the mechanism but also as a therapeutic strategy. Acknowledgments. This work was supported NIH Research Grants SC1GM082325, R25 HL003679, 2G12RR003048, 8G12MD007597, K25GM097501 and 1P30HL107253. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 2014 ◽  
pp. 1-8 ◽  
Author(s):  
Sergei Nekhai ◽  
Michael Petukhov ◽  
Denitra Breuer

HIV-1 transcription is regulated by CDK9/cyclin T1, which, unlike a typical cell cycle-dependent kinase, is regulated by associating with 7SK small nuclear ribonuclear protein complex (snRNP). While the protein components of this complex are well studied, the mechanism of the complex formation is still not fully understood. The association of CDK9/cyclin T1 with 7SK snRNP is, in part, regulated by a reversible CDK9 phosphorylation. Here, we present a comprehensive review of the kinases and phosphatases involved in CDK9 phosphorylation and discuss their role in regulation of HIV-1 replication and potential for being targeted for drug development. We propose a novel pathway of HIV-1 transcription regulation via CDK9 Ser-90 phosphorylation by CDK2 and CDK9 Ser-175 dephosphorylation by protein phosphatase-1.


2014 ◽  
Vol 58 (11) ◽  
pp. 6558-6571 ◽  
Author(s):  
Namita Kumari ◽  
Sergey Iordanskiy ◽  
Dmytro Kovalskyy ◽  
Denitra Breuer ◽  
Xiaomei Niu ◽  
...  

ABSTRACTHIV-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.


2010 ◽  
Vol 84 (23) ◽  
pp. 12300-12314 ◽  
Author(s):  
Hanna-Mari Tervo ◽  
Oliver T. Keppler

ABSTRACT An immunocompetent, permissive, small-animal model would be valuable for the study of human immunodeficiency virus type 1 (HIV-1) pathogenesis and for the testing of drug and vaccine candidates. However, the development of such a model has been hampered by the inability of primary rodent cells to efficiently support several steps of the HIV-1 replication cycle. Although transgenesis of the HIV receptor complex and human cyclin T1 have been beneficial, additional late-phase blocks prevent robust replication of HIV-1 in rodents and limit the range of in vivo applications. In this study, we explored the HIV-1 susceptibility of rabbit primary T cells and macrophages. Envelope-specific and coreceptor-dependent entry of HIV-1 was achieved by expressing human CD4 and CCR5. A block of HIV-1 DNA synthesis, likely mediated by TRIM5, was overcome by limited changes to the HIV-1 gag gene. Unlike with mice and rats, primary cells from rabbits supported the functions of the regulatory viral proteins Tat and Rev, Gag processing, and the release of HIV-1 particles at levels comparable to those in human cells. While HIV-1 produced by rabbit T cells was highly infectious, a macrophage-specific infectivity defect became manifest by a complex pattern of mutations in the viral genome, only part of which were deamination dependent. These results demonstrate a considerable natural HIV-1 permissivity of the rabbit species and suggest that receptor complex transgenesis combined with modifications in gag and possibly vif of HIV-1 to evade species-specific restriction factors might render lagomorphs fully permissive to infection by this pathogenic human lentivirus.


1995 ◽  
Vol 23 (20) ◽  
pp. 4050-4054 ◽  
Author(s):  
Masatomo Yonaha ◽  
Taku Chibazakura ◽  
Shigetaka Kitajima ◽  
Yukio Yasukochi

2016 ◽  
Vol 3 (12) ◽  
pp. 160578 ◽  
Author(s):  
Mohammad Soltani ◽  
Abhyudai Singh

Expression of many genes varies as a cell transitions through different cell-cycle stages. How coupling between stochastic expression and cell cycle impacts cell-to-cell variability (noise) in the level of protein is not well understood. We analyse a model where a stable protein is synthesized in random bursts, and the frequency with which bursts occur varies within the cell cycle. Formulae quantifying the extent of fluctuations in the protein copy number are derived and decomposed into components arising from the cell cycle and stochastic processes. The latter stochastic component represents contributions from bursty expression and errors incurred during partitioning of molecules between daughter cells. These formulae reveal an interesting trade-off: cell-cycle dependencies that amplify the noise contribution from bursty expression also attenuate the contribution from partitioning errors. We investigate the existence of optimum strategies for coupling expression to the cell cycle that minimize the stochastic component. Intriguingly, results show that a zero production rate throughout the cell cycle, with expression only occurring just before cell division, minimizes noise from bursty expression for a fixed mean protein level. By contrast, the optimal strategy in the case of partitioning errors is to make the protein just after cell division. We provide examples of regulatory proteins that are expressed only towards the end of the cell cycle, and argue that such strategies enhance robustness of cell-cycle decisions to the intrinsic stochasticity of gene expression.


2021 ◽  
Author(s):  
Siv Anita Hegre ◽  
Helle Samdal ◽  
Antonin Klima ◽  
Endre B. Stovner ◽  
Kristin G. Nørsett ◽  
...  

AbstractProper regulation of the cell cycle is necessary for normal growth and development of all organisms. Conversely, altered cell cycle regulation often underlies proliferative diseases such as cancer. Long non-coding RNAs (lncRNAs) are recognized as important regulators of gene expression and are often found dysregulated in diseases, including cancers. However, identifying lncRNAs with cell cycle functions is challenging due to their often low and cell-type specific expression. We present a highly effective method that analyses changes in promoter activity, transcription, and RNA levels for identifying genes enriched for cell cycle functions. Specifically, by combining RNA sequencing with ChIP sequencing through the cell cycle of synchronized human keratinocytes, we identified 1009 genes with cell cycle-dependent expression and correlated changes in RNA polymerase II occupancy or promoter activity as measured by histone 3 lysine 4 trimethylation (H3K4me3). These genes were highly enriched for genes with known cell cycle functions and included 59 lncRNAs. We selected four of these lncRNAs – AC005682.5, RP11-132A1.4, ZFAS1, and EPB41L4A-AS1 – for further experimental validation and found that knockdown of each of the four lncRNAs affected cell cycle phase distributions and reduced proliferation in multiple cell lines. These results show that many genes with cell cycle functions have concomitant cell-cycle dependent changes in promoter activity, transcription, and RNA levels and support that our multi-omics method is well suited for identifying lncRNAs involved in the cell cycle.


Sign in / Sign up

Export Citation Format

Share Document