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











Publication year range
1.
PLoS Biol ; 20(9): e3001777, 2022 09.
Article in English | MEDLINE | ID: mdl-36112666

ABSTRACT

Wound healing in the skin is a complex physiological process that is a product of a cell state transition from homeostasis to repair. Mechanical cues are increasingly being recognized as important regulators of cellular reprogramming, but the mechanism by which it is translated to changes in gene expression and ultimately cellular behavior remains largely a mystery. To probe the molecular underpinnings of this phenomenon further, we used the down-regulation of caspase-8 as a biomarker of a cell entering the wound healing program. We found that the wound-induced release of tension within the epidermis leads to the alteration of gene expression via the nuclear translocation of the DNA methyltransferase 3A (DNMT3a). This enzyme then methylates promoters of genes that are known to be down-regulated in response to wound stimuli as well as potentially novel players in the repair program. Overall, these findings illuminate the convergence of mechanical and epigenetic signaling modules that are important regulators of the transcriptome landscape required to initiate the tissue repair process in the differentiated layers of the epidermis.


Subject(s)
Cues , Wound Healing , Biomarkers , Caspase 8 , Epigenesis, Genetic , Wound Healing/genetics
2.
Stem Cell Res ; 37: 101444, 2019 05.
Article in English | MEDLINE | ID: mdl-31075691

ABSTRACT

Human pluripotent stem cells (hPSCs) acquire changes at the genomic level upon proliferation and differentiation (Peterson and Loring, 2014). Studies from International Stem Cell Initiative and independent laboratories identified a copy number variant (CNV) in hES cell lines displaying a normal karyotype, which provided a selective advantage to hES cells in culture. In our laboratory we have identified variant H9-hESC (derived from H9-hESC) with normal karyotype, pluripotency expression, differentiation profile but with altered traits of high cell survival and low E-CADHERIN expression.


Subject(s)
Biomedical Research/statistics & numerical data , Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Teratoma/pathology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred NOD , Mice, SCID
3.
EMBO Rep ; 20(5)2019 05.
Article in English | MEDLINE | ID: mdl-30858340

ABSTRACT

Euchromatic histone methyltransferases (EHMTs), members of the KMT1 family, methylate histone and non-histone proteins. Here, we uncover a novel role for EHMTs in regulating heterochromatin anchorage to the nuclear periphery (NP) via non-histone methylation. We show that EHMTs methylate and stabilize LaminB1 (LMNB1), which associates with the H3K9me2-marked peripheral heterochromatin. Loss of LMNB1 methylation or EHMTs abrogates heterochromatin anchorage at the NP We further demonstrate that the loss of EHMTs induces many hallmarks of aging including global reduction of H3K27methyl marks and altered nuclear morphology. Consistent with this, we observe a gradual depletion of EHMTs, which correlates with loss of methylated LMNB1 and peripheral heterochromatin in aging human fibroblasts. Restoration of EHMT expression reverts peripheral heterochromatin defects in aged cells. Collectively, our work elucidates a new mechanism by which EHMTs regulate heterochromatin domain organization and reveals their impact on fundamental changes associated with the intrinsic aging process.


Subject(s)
Cell Nucleus/metabolism , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lamin Type B/metabolism , Aging/metabolism , Cell Line , HEK293 Cells , Humans , Methylation
4.
Nature ; 560(7719): E32, 2018 08.
Article in English | MEDLINE | ID: mdl-30042505

ABSTRACT

In this Article, there were duplicated empty lanes in Supplementary Figs. 2e and 3b. The corrected figures are presented in the Supplementary Information to the accompanying Amendment. The original Article has not been corrected.

5.
Dis Model Mech ; 8(10): 1295-309, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26398939

ABSTRACT

Medulloblastoma (MB) is the most common malignant primary pediatric brain tumor and is currently divided into four subtypes based on different genomic alterations, gene expression profiles and response to treatment: WNT, Sonic Hedgehog (SHH), Group 3 and Group 4. This extensive heterogeneity has made it difficult to assess the functional relevance of genes to malignant progression. For example, expression of the transcription factor Orthodenticle homeobox2 (OTX2) is frequently dysregulated in multiple MB variants; however, its role may be subtype specific. We recently demonstrated that neural precursors derived from transformed human embryonic stem cells (trans-hENs), but not their normal counterparts (hENs), resemble Groups 3 and 4 MB in vitro and in vivo. Here, we tested the utility of this model system as a means of dissecting the role of OTX2 in MB using gain- and loss-of-function studies in hENs and trans-hENs, respectively. Parallel experiments with MB cells revealed that OTX2 exerts inhibitory effects on hEN and SHH MB cells by regulating growth, self-renewal and migration in vitro and tumor growth in vivo. This was accompanied by decreased expression of pluripotent genes, such as SOX2, and was supported by overexpression of SOX2 in OTX2+ SHH MB and hENs that resulted in significant rescue of self-renewal and cell migration. By contrast, OTX2 is oncogenic and promotes self-renewal of trans-hENs and Groups 3 and 4 MB independent of pluripotent gene expression. Our results demonstrate a novel role for OTX2 in self-renewal and migration of hENs and MB cells and reveal a cell-context-dependent link between OTX2 and pluripotent genes. Our study underscores the value of human embryonic stem cell derivatives as alternatives to cell lines and heterogeneous patient samples for investigating the contribution of key developmental regulators to MB progression.


Subject(s)
Human Embryonic Stem Cells/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Neural Stem Cells/metabolism , Otx Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Self Renewal , Cell Survival , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Medulloblastoma/genetics , Models, Biological , Neural Stem Cells/pathology , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Transcriptome/genetics
6.
Sci Rep ; 5: 8229, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25648270

ABSTRACT

Factor induced reprogramming of fibroblasts is an orchestrated but inefficient process. At the epigenetic level, it results in drastic chromatin changes to erase the existing somatic "memory" and to establish the pluripotent state. Accordingly, alterations of chromatin regulators including Ezh2 influence iPSC generation. While the role of individual transcription factors in resetting the chromatin landscape during iPSC generation is increasingly evident, their engagement with chromatin modulators remains to be elucidated. In the current study, we demonstrate that histone methyl transferase activity of Ezh2 is required for mesenchymal to epithelial transition (MET) during human iPSC generation. We show that the H3K27me3 activity favors induction of pluripotency by transcriptionally targeting the TGF-ß signaling pathway. We also demonstrate that the Ezh2 negatively regulates the expression of pro-EMT miRNA's such as miR-23a locus during MET. Unique association of Ezh2 with c-Myc was required to silence the aforementioned circuitry. Collectively, our findings provide a mechanistic understanding by which Ezh2 restricts the somatic programme during early phase of cellular reprogramming and establish the importance of Ezh2 dependent H3K27me3 activity in transcriptional and miRNA modulation during human iPSC generation.


Subject(s)
Cellular Reprogramming , Histones/metabolism , Induced Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Cell Differentiation/genetics , Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , MicroRNAs/genetics , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/genetics , Protein Binding , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Transforming Growth Factor beta/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Blood ; 121(9): 1543-52, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23293081

ABSTRACT

Programs that control early lineage fate decisions and transitions from embryonic to adult human cell types during development are poorly understood. Using human pluripotent stem cells (hPSCs), in the present study, we reveal reduction of Hedgehog (Hh) signaling correlates to developmental progression of hematopoiesis throughout human ontogeny. Both chemical- and gene-targeting­mediated inactivation of Hh signaling augmented hematopoietic fate and initiated transitions from embryonic to adult hematopoiesis, as measured by globin regulation in hPSCs. Inhibition of the Hh pathway resulted in truncation of Gli3 to its repressor, Gli3R, and was shown to be necessary and sufficient for initiating this transition. Our results reveal an unprecedented role for Hh signaling in the regulation of adult hematopoietic specification, thereby demonstrating the ability to modulate the default embryonic programs of hPSCs.


Subject(s)
Hedgehog Proteins/genetics , Hematopoiesis/genetics , Kruppel-Like Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Blood Cells/metabolism , Blood Cells/physiology , Cell Differentiation/genetics , Cells, Cultured , Down-Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Microarray Analysis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcriptome , Zinc Finger Protein Gli3
8.
Cell Cycle ; 11(13): 2411-2, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22713241
9.
Cell Stem Cell ; 9(1): 24-36, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21726831

ABSTRACT

Human embryonic stem cells (hESCs) expressing pluripotency markers are assumed to possess equipotent developmental potential. However, disparate responses to differentiation stimuli functionally illustrate that hESCs generate a spectrum of differentiated cell types, suggestive of lineage bias. Here, we reveal specific cell surface markers that allow subfractionation of hESCs expressing hallmark markers of pluripotency. By direct de novo isolation of these subsets, we demonstrate that propensities for lineage differentiation are balanced with reduced clonogenic self-renewal. Histone modification marks of gene loci associated with pluripotency versus lineage specificity predicted cell fate potential of these subfractions, thereby supporting the absence of uniform bivalency as a molecular paradigm to describe cell fate determination of pluripotent cells. Our study reveals that cell fate potential is encoded within cells comprising hESC cultures, highlighting them as a means to understand the mechanisms of lineage specification of pluripotent cells.


Subject(s)
Cell Lineage , Histones/metabolism , Pluripotent Stem Cells/cytology , Protein Processing, Post-Translational , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Clone Cells , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gangliosides/metabolism , Hematopoiesis , Humans , Mice , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism
10.
Biochim Biophys Acta ; 1815(1): 1-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20709157

ABSTRACT

Changes in the composition of nuclear matrix associated proteins contribute to alterations in nuclear structure, one of the major phenotypes of malignant cancer cells. The malignancy-induced changes in this structure lead to alterations in chromatin folding, the fidelity of genome replication and gene expression programs. The nuclear matrix forms a scaffold upon which the chromatin is organized into periodic loop domains called matrix attachment regions (MAR) by binding to various MAR binding proteins (MARBPs). Aberrant expression of MARBPs modulates the chromatin organization and disrupt transcriptional network that leads to oncogenesis. Dysregulation of nuclear matrix associated MARBPs has been reported in different types of cancers. Some of these proteins have tumor specific expression and are therefore considered as promising diagnostic or prognostic markers in few cancers. SMAR1 (scaffold/matrix attachment region binding protein 1), is one such nuclear matrix associated protein whose expression is drastically reduced in higher grades of breast cancer. SMAR1 gene is located on human chromosome 16q24.3 locus, the loss of heterozygosity (LOH) of which has been reported in several types of cancers. This review elaborates on the multiple roles of nuclear matrix associated protein SMAR1 in regulating various cellular target genes involved in cell growth, apoptosis and tumorigenesis.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation , Homeostasis , Neoplasms/etiology , Nuclear Proteins/physiology , Animals , Apoptosis , Cell Cycle , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Genes, bcl-1 , Humans , Neoplasm Invasiveness , Nuclear Proteins/genetics , Signal Transduction , Transforming Growth Factor beta/physiology
11.
Nature ; 468(7323): 521-6, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-21057492

ABSTRACT

As is the case for embryo-derived stem cells, application of reprogrammed human induced pluripotent stem cells is limited by our understanding of lineage specification. Here we demonstrate the ability to generate progenitors and mature cells of the haematopoietic fate directly from human dermal fibroblasts without establishing pluripotency. Ectopic expression of OCT4 (also called POU5F1)-activated haematopoietic transcription factors, together with specific cytokine treatment, allowed generation of cells expressing the pan-leukocyte marker CD45. These unique fibroblast-derived cells gave rise to granulocytic, monocytic, megakaryocytic and erythroid lineages, and demonstrated in vivo engraftment capacity. We note that adult haematopoietic programs are activated, consistent with bypassing the pluripotent state to generate blood fate: this is distinct from haematopoiesis involving pluripotent stem cells, where embryonic programs are activated. These findings demonstrate restoration of multipotency from human fibroblasts, and suggest an alternative approach to cellular reprogramming for autologous cell-replacement therapies that avoids complications associated with the use of human pluripotent stem cells.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Fibroblasts/cytology , Hematopoietic Stem Cells/cytology , Stem Cells/cytology , Dermis/cytology , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
12.
EMBO J ; 29(8): 1401-11, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20300060

ABSTRACT

Polycomb (PcG) and Trithorax (TrxG) group proteins act antagonistically to establish tissue-specific patterns of gene expression. The PcG protein Ezh2 facilitates repression by catalysing histone H3-Lys27 trimethylation (H3K27me3). For expression, H3K27me3 marks are removed and replaced by TrxG protein catalysed histone H3-Lys4 trimethylation (H3K4me3). Although H3K27 demethylases have been identified, the mechanism by which these enzymes are targeted to specific genomic regions to remove H3K27me3 marks has not been established. Here, we demonstrate a two-step mechanism for UTX-mediated demethylation at muscle-specific genes during myogenesis. Although the transactivator Six4 initially recruits UTX to the regulatory region of muscle genes, the resulting loss of H3K27me3 marks is limited to the region upstream of the transcriptional start site. Removal of the repressive H3K27me3 mark within the coding region then requires RNA Polymerase II (Pol II) elongation. Interestingly, blocking Pol II elongation on transcribed genes leads to increased H3K27me3 within the coding region, and formation of bivalent (H3K27me3/H3K4me3) chromatin domains. Thus, removal of repressive H3K27me3 marks by UTX occurs through targeted recruitment followed by spreading across the gene.


Subject(s)
Histones/metabolism , Muscle Development , Nuclear Proteins/metabolism , Animals , Cell Line , Creatine Kinase/metabolism , Genes , Histone Demethylases/metabolism , Homeodomain Proteins/metabolism , Methylation , Mice , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/genetics , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/metabolism , Trans-Activators/metabolism
13.
Proc Natl Acad Sci U S A ; 107(9): 4230-5, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20160104

ABSTRACT

Caspase 3 is required for the differentiation of a wide variety of cell types, yet it remains unclear how this apoptotic protein could promote such a cell-fate decision. Caspase signals often result in the activation of the specific nuclease caspase-activated DNase (CAD), suggesting that cell differentiation may be dependent on a CAD-mediated modification in chromatin structure. In this study, we have investigated if caspase 3/CAD plays a role in initiating the DNA strand breaks that are known to occur during the terminal differentiation of skeletal muscle cells. Here, we show that inhibition of caspase 3 or reduction of CAD expression leads to a dramatic loss of strand-break formation and a block in the myogenic program. Caspase-dependent induction of differentiation results in CAD targeting of the p21 promoter, and loss of caspase 3 or CAD leads to a significant down-regulation in p21 expression. These results show that caspase 3/CAD promotes cell differentiation by directly modifying the DNA/nuclear microenvironment, which enhances the expression of critical regulatory genes.


Subject(s)
Caspase 3/metabolism , Cell Differentiation , DNA Damage , Deoxyribonucleases/metabolism , Animals , Base Sequence , Cell Line , Chromatin Immunoprecipitation , DNA Primers , Enzyme Activation , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism
14.
Nat Protoc ; 3(3): 398-409, 2008.
Article in English | MEDLINE | ID: mdl-18323811

ABSTRACT

Chromatin immunoprecipitation (ChIP) is routinely used to examine epigenetic modification of histones at specific genomic locations. However, covalent modifications of histone tails can serve as docking sites for chromatin regulatory factors. As such, association of these regulatory factors with chromatin could cause steric hindrance for antibody recognition, resulting in an underestimation of the relative enrichment of a given histone modification at specific loci. To overcome this problem, we have developed a native ChIP protocol to study covalent modification of histones that takes advantage of hydroxyapatite (HAP) chromatography to wash away chromatin-associated proteins before the immunoprecipitation of nucleosomes. This fast and simple procedure consists of five steps: nuclei isolation from cultured cells; fragmentation of chromatin using MNase; purification of nucleosomes using HAP; immunoprecipitation of modified nucleosomes; and qPCR analysis of DNA associated with modified histones. Nucleosomes prepared in this manner are free of contaminating proteins and permit an accurate evaluation of relative abundance of different covalent histone modifications at specific genomic loci. Completion of this protocol requires approximately 1.5 d.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatography/methods , Nucleosomes/immunology , Durapatite/chemistry , Epigenesis, Genetic/genetics , Epitopes/analysis , Nucleosomes/genetics , Polymerase Chain Reaction/methods
15.
Nat Struct Mol Biol ; 14(12): 1150-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18026121

ABSTRACT

Cell-specific patterns of gene expression are established through the antagonistic functions of trithorax group (TrxG) and Polycomb group (PcG) proteins. Several muscle-specific genes have previously been shown to be epigenetically marked for repression by PcG proteins in muscle progenitor cells. Here we demonstrate that these developmentally regulated genes become epigenetically marked for gene expression (trimethylated on histone H3 Lys4, H3K4me3) during muscle differentiation through specific recruitment of Ash2L-containing methyltransferase complexes. Targeting of Ash2L to specific genes is mediated by the transcriptional regulator Mef2d. Furthermore, this interaction is modulated during differentiation through activation of the p38 MAPK signaling pathway via phosphorylation of Mef2d. Thus, we provide evidence that signaling pathways regulate the targeting of TrxG-mediated epigenetic modifications at specific promoters during cellular differentiation.


Subject(s)
Cell Differentiation , Chromosomal Proteins, Non-Histone/genetics , Methyltransferases/metabolism , Repressor Proteins/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , Mice , Muscles/enzymology , Muscles/metabolism , Polycomb-Group Proteins , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
16.
Nucleic Acids Res ; 35(18): 6004-16, 2007.
Article in English | MEDLINE | ID: mdl-17726044

ABSTRACT

Prostaglandins are anticancer agents known to inhibit tumor cell proliferation both in vitro and in vivo by affecting the mRNA stability. Here we report that a MAR-binding protein SMAR1 is a target of Prostaglandin A2 (PGA2) induced growth arrest. We identify a regulatory mechanism leading to stabilization of SMAR1 transcript. Our results show that a minor stem and loop structure present in the 5' UTR of SMAR1 (1-UTR) is critical for nucleoprotein complex formation that leads to SMAR1 stabilization in response to PGA2. This results in an increased SMAR1 transcript and altered protein levels, that in turn causes downregulation of Cyclin D1 gene, essential for G1/S phase transition. We also provide evidence for the presence of a variant 5' UTR SMAR1 (17-UTR) in breast cancer-derived cell lines. This form lacks the minor stem and loop structure required for mRNA stabilization in response to PGA2. As a consequence of this, there is a low level of endogenous tumor suppressor protein SMAR1 in breast cancer-derived cell lines. Our studies provide a mechanistic insight into the regulation of tumor suppressor protein SMAR1 by a cancer therapeutic PGA2, that leads to repression of Cyclin D1 gene.


Subject(s)
5' Untranslated Regions/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Prostaglandins A/pharmacology , RNA Stability/drug effects , RNA, Messenger/metabolism , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cyclin D , Cyclins/metabolism , DNA-Binding Proteins/biosynthesis , Humans , Molecular Sequence Data , Nuclear Proteins/biosynthesis , Nucleic Acid Conformation
17.
Mol Cell Biol ; 25(19): 8415-29, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16166625

ABSTRACT

Matrix attachment region binding proteins have been shown to play an important role in gene regulation by altering chromatin in a stage- and tissue-specific manner. Our previous studies report that SMAR1, a matrix-associated protein, regresses B16-F1-induced tumors in mice. Here we show SMAR1 targets the cyclin D1 promoter, a gene product whose dysregulation is attributed to breast malignancies. Our studies reveal that SMAR1 represses cyclin D1 gene expression, which can be reversed by small interfering RNA specific to SMAR1. We demonstrate that SMAR1 interacts with histone deacetylation complex 1, SIN3, and pocket retinoblastomas to form a multiprotein repressor complex. This interaction is mediated by the SMAR1(160-350) domain. Our data suggest SMAR1 recruits a repressor complex to the cyclin D1 promoter that results in deacetylation of chromatin at that locus, which spreads to a distance of at least the 5 kb studied upstream of the cyclin D1 promoter. Interestingly, we find that the high induction of cyclin D1 in breast cancer cell lines can be correlated to the decreased levels of SMAR1 in these lines. Our results establish the molecular mechanism exhibited by SMAR1 to regulate cyclin D1 by modification of chromatin.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin D1/chemistry , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Binding Sites , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Size , Chromatin/metabolism , Chromatin Immunoprecipitation , Cyclin D1/metabolism , DNA Primers/chemistry , G1 Phase , Gene Expression Regulation, Neoplastic , Glutathione Transferase/metabolism , Histone Deacetylase 1 , Histones/metabolism , Humans , Immunoblotting , Immunoprecipitation , Luciferases/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Retinoblastoma/metabolism , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Sin3 Histone Deacetylase and Corepressor Complex , Time Factors
18.
Biochem Biophys Res Commun ; 329(2): 706-12, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15737643

ABSTRACT

The HIV-1 transactivator Tat performs various viral and cellular functions. Primarily, it induces processive transcription from the HIV-1 LTR promoter. However, Tat secreted from infected cells is known to activate uninfected lymphocytes through receptors. To further delineate the specific target genes, extracellular Tat was expressed on the cell membrane of stimulator cells and co-cultured with human PBMCs. Along with induced CD4(+) T cell proliferation and IFN-gamma secretion, there was strong upregulation of T-bet expression which is majorly implicated in generating T(H)1 type of immune response. To further delineate the effect of extracellular Tat on HIV replication, both p24 analysis and in vivo GFP expression were performed. There was a significant inhibition of HIV-1 replication in human CEM-GFP cell line and hPBMCs. Thus, for the first time we report that apart from its transactivation activity, extracellular Tat acts as a costimulatory molecule that affects viral replication by modulating host immune response through induction of T-bet expression and IFN-gamma secretion.


Subject(s)
Gene Products, tat/immunology , HIV-1/growth & development , HIV-1/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Th1 Cells/immunology , Transcription Factors/immunology , Coculture Techniques , Humans , T-Box Domain Proteins , Virus Replication/immunology , tat Gene Products, Human Immunodeficiency Virus
19.
Biochem Biophys Res Commun ; 322(2): 672-7, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15325282

ABSTRACT

Matrix attachment regions (MARs) are cis regulatory elements that modulate gene expression in a tissue and cell stage specific manner. Recent reports show that viral integration within the genome takes place at nonrandom active genes. We have checked for the presence of MARs in the vicinity of the reported 524 HIV-1 integration sites. Our studies show that in 92.5% cases, MARs flank the integration sites. Similarly, for adeno-associated virus, two potential MARs were present next to the integration site on the human chromosome. Earlier we have shown that short MAR sequences present upstream of HIV-1 LTR promote processive transcription at a distance. Here, using a well-studied IgH-MAR and another potential MAR from p53 promoter, we demonstrate that MARs alone can act as promoters. Thus, we propose that MAR elements near the HIV-1 integration sites can act as potential promoters, which may facilitate proviral integration and transcription.


Subject(s)
HIV-1/genetics , Matrix Attachment Regions/genetics , Promoter Regions, Genetic , Virus Integration/genetics , Chromosome Mapping , Dependovirus/genetics , Humans , Immunoglobulin Heavy Chains/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics
20.
Nucleic Acids Res ; 31(12): 3248-56, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12799452

ABSTRACT

The chromatin environment and the sites of integration in the host genome are critical determinants of human immunodeficiency virus (HIV) transcription and replication. Depending on the chromosomal location of provirus integration within the genome, HIV-1 long terminal repeat (LTR)-mediated transcription may vary from 0- to 70-fold. Cis-elements such as topoisomerase II cleavage sites, Alu repeats and matrix attachment regions (MARs) are thought to be targets for retroviral integration. Here we show that a novel MAR sequence from the T-cell receptor beta locus (MARbeta) and the IgH MAR mediate transcriptional augmentation when placed upstream of the HIV-1 LTR promoter. The effect of transcriptional augmentation is seen in both transient and stable transfection, indicating its effect even upon integration in the genome. MAR-mediated transcriptional elevation is independent of Tat, and occurs synergistically in the presence of Tat. Further, we show that MAR-mediated transcriptional elevation is specific to the HIV-1 LTR and the Moloney murine leukemia virus LTR promoter. In a transient transfection assay using over-expressed IkappaB, the inhibitor of NF-kappaB, we show that MAR-induced processive transcription is NF-kappaB dependent, signifying the role of local enhancers within the LTR promoter. Furthermore, by RNase protection experiments using proximal and distal probes, we show that MAR-mediated transcriptional upregulation is more prominent at the distal rather than the proximal end, thus indicating the potential role of MARs in promoting elongation.


Subject(s)
HIV Long Terminal Repeat , HIV-1/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcriptional Activation , Animals , CHO Cells , Cell Line , Cricetinae , Gene Expression Regulation, Viral , Gene Products, tat/physiology , Genes, T-Cell Receptor beta , Immunoglobulin Heavy Chains/genetics , NF-kappa B/physiology , Nuclear Matrix/metabolism , Peptide Chain Elongation, Translational , Sequence Deletion , Virus Integration , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL