Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Methods ; 121-122: 45-54, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28499832

ABSTRACT

The advent of programmable nucleases such as ZFNs, TALENs and CRISPR/Cas9 has brought the power of genetic manipulation to widely used model systems. In mammalian cells, nuclease-mediated DNA double strand break is mainly repaired through the error-prone non-homologous end-joining (NHEJ) repair pathway, eventually leading to accumulation of small deletions or insertions (indels) that can inactivate gene function. However, due to the variable size of the indels and the polyploid status of many cell lines (e.g., cancer-derived cells), obtaining a knockout usually requires lengthy screening and characterization procedures. Given the more precise type of modifications that can be introduced upon homology-directed repair (HDR), we have developed HDR-based gene-targeting strategies that greatly facilitate the process of knockout generation in cell lines. To generate reversible knockouts (R-KO), a selectable promoter-less STOP cassette is inserted in an intron, interrupting transcription. Loss-of-function can be validated by RT-qPCR and is removable, enabling subsequent restoration of gene function. A variant of the R-KO procedure can be used to introduce point mutations. To generate constitutive knockouts (C-KO), an exon is targeted, which makes use of HDR-based gene disruption together with NHEJ-induced indels on non-HDR targeted allele(s). Hence the C-KO procedure greatly facilitates simultaneous inactivation of multiple alleles. Overall these genome-editing tools offer superior precision and efficiency for functional genetic approaches. We provide detailed protocols guiding in the design of targeting vectors and in the analysis and validation of gene targeting experiments.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Endonucleases/genetics , Gene Editing/methods , Gene Knockout Techniques , Gene Transfer Techniques , RNA, Guide, Kinetoplastida/genetics , Animals , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Clone Cells , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Endonucleases/metabolism , Exons , Gene Targeting/methods , Genome , HEK293 Cells , Humans , Introns , Mice , NIH 3T3 Cells , Point Mutation , RNA, Guide, Kinetoplastida/metabolism , Recombinational DNA Repair , Transcription, Genetic
2.
J Mol Biol ; 429(13): 1978-1993, 2017 06 30.
Article in English | MEDLINE | ID: mdl-27742591

ABSTRACT

Genome sequencing of large cohorts of tumors has revealed that mutations in genes encoding chromatin regulators are frequent in cancer. However, the precise contribution of these mutations to tumor development often remains elusive. Here, we review the current knowledge concerning the alterations of the Polycomb machinery in cancer, with a particular focus on the Polycomb repressive complex 2 (PRC2), a key chromatin modifier involved in the maintenance of transcriptional silencing. A broad variety of alterations can impair PRC2 activity; yet, overall, only one type of alteration is found in a given class of tumor. We discuss the potential impact of the various types of PRC2 alterations on gene expression. We propose that the distinct set of genes regulated by PRC2, depending on tumor etiology, constrain the type of alteration of PRC2 that can fuel tumor development. Beyond this specificity, we propose that PRC2 and, more generally, chromatin regulators act as gatekeepers of transcriptional integrity, a role that often confers a tumor-suppressive function.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Neoplasms/pathology , Neoplasms/physiopathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Humans
3.
Cell Cycle ; 15(17): 2256-62, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27419533

ABSTRACT

EZH2, the main catalytic component of the Polycomb Repressive Complex 2 (PRC2) is apparently upregulated in most solid tumors. Furthermore its expression generally associates with poor prognosis. It was proposed that this correlation reflects a causal event, EZH2 mediating the silencing of key tumor suppressor loci. In contrast, we recently showed that EZH2 is dispensable for solid tumor development and that its elevated expression reflects the abnormally high proliferation rate of cancer cells. Here, we investigate the functional association between EZH2 expression and silencing of key tumor suppressor loci and further illustrate the confounding effect of proliferation on EZH2's association to outcome.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Silencing , Genes, Tumor Suppressor , Neoplasms/genetics , Animals , Biomarkers, Tumor/metabolism , Cell Line, Transformed , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Mice , Models, Biological , Neoplasms/pathology , Treatment Outcome
4.
Mol Cell Biol ; 33(24): 5005-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144980

ABSTRACT

X chromosome inactivation is a remarkable example of chromosome-wide gene silencing and facultative heterochromatin formation. Numerous histone posttranslational modifications, including H3K9me2 and H3K27me3, accompany this process, although our understanding of the enzymes that lay down these marks and the factors that bind to them is still incomplete. Here we identify Cdyl, a chromodomain-containing transcriptional corepressor, as a new chromatin-associated protein partner of the inactive X chromosome (Xi). Using mouse embryonic stem cell lines with mutated histone methyltransferase activities, we show that Cdyl relies on H3K9me2 for its general association with chromatin in vivo. For its association with Xi, Cdyl requires the process of differentiation and the presence of H3K9me2 and H3K27me3, which both become chromosomally enriched following Xist RNA coating. We further show that the removal of the PRC2 component Eed and subsequent loss of H3K27me3 lead to a reduction of both Cdyl and H3K9me2 enrichment on inactive Xi. Finally, we show that Cdyl associates with the H3K9 histone methyltransferase G9a and the MGA protein, both of which are also found on Xi. We propose that the combination of H3K9me2 and H3K27me3 recruits Cdyl to Xi, and this, in turn, may facilitate propagation of the H3K9me2 mark by anchoring G9a.


Subject(s)
Embryonic Stem Cells/physiology , Histones/metabolism , Proteins/metabolism , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Co-Repressor Proteins , Epigenesis, Genetic , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Histone Acetyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Hydro-Lyases , Methylation , Mice , Polycomb Repressive Complex 1/metabolism , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Stability , Tretinoin/physiology , X Chromosome/genetics
5.
Article in English | MEDLINE | ID: mdl-21502408

ABSTRACT

Chromatin affects many, if not all aspects, of nuclear organization and function. For this reason, we have focused our attention on elucidating some of the basic mechanisms regulating the formation and maintenance of chromatin, specifically concerning Polycomb repressive complex 2 (PRC2) and PR-Set7. PRC2 is responsible for catalyzing trimethylation of lysine 27 of histone H3 and thus has a critical role in the formation of facultative heterochromatin. PR-Set7 is responsible for catalyzing monomethylation of lysine 20 of histone H4 and is required for proper cell cycle progression and DNA damage response. We have also expanded our work to establish novel techniques and approaches to determine how chromatin is spatially regulated within the nuclear landscape.


Subject(s)
Chromatin/metabolism , Animals , Cell Cycle , DNA Damage , Heterochromatin/metabolism , Humans , Polycomb-Group Proteins , Protein Structure, Tertiary , Repressor Proteins/metabolism
6.
Oncogene ; 25(12): 1799-806, 2006 Mar 16.
Article in English | MEDLINE | ID: mdl-16158045

ABSTRACT

The proliferative action of ERalpha largely accounts for the carcinogenic activity of estrogens. By contrast, recent data show that ERbeta displays tumor-suppressor properties, thus supporting the interest to identify compounds that could increase its activity. Here, we show that histone deacetylase inhibitors (HDI) upregulated ERbeta protein levels, whereas it decreased ERalpha expression. Part of this regulation took place at the mRNA level through a mechanism independent of de novo protein synthesis. In addition, we found that, in various cancer cells, the treatment with different HDI enhanced the ligand-dependent activity of ERbeta more strongly than that of ERalpha. On the other hand, in MDA-MB231 and HeLa cells, the expression of ERs modified the transcriptional response to HDI. The use of deletion mutants of both receptors demonstrated that AF1 domain of the receptors was required. Finally, we show that ERbeta expression led to a dramatic increased in the antiproliferative activity of HDI, which correlated with a modification of the transcription of genes involved in cell cycle control by HDI. Altogether, these data demonstrate that the interference of ERbeta and HDAC on the control of transcription and cell proliferation constitute a promising approach for cancer therapy.


Subject(s)
Enzyme Inhibitors/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Histone Deacetylases/metabolism , Transcription, Genetic/drug effects , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , HeLa Cells , Histone Deacetylases/drug effects , Humans , Polymerase Chain Reaction , RNA, Messenger
7.
Endocrinology ; 146(2): 760-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15528301

ABSTRACT

Fibulin-1 is an extracellular matrix protein overexpressed in epithelial ovarian and breast cancers. In estrogen receptor (ER)-positive ovarian and breast cancer cell lines, fibulin-1 mRNA levels are markedly increased by estrogens. Transfection experiments using fibulin-1 promoter constructs indicate that 17beta-estradiol (E2) increases fibulin-1 gene transcription and that ERalpha is more potent than ERbeta to mediate E2 regulation of the transfected fibulin-1 promoter. Using SL2 cells devoid of specificity protein 1 (Sp1) and site-directed mutagenesis of GC boxes, we evidenced that the E2 regulation occurs through a proximal specificity protein 1 binding site. In addition, we show that fibulin-1C and -1D mRNAs, the two major fibulin-1 splicing variants, are differentially induced by E2. The induction of both mRNAs variants is direct and independent of a newly synthesized protein intermediate. Interestingly, actinomycin D chase experiments demonstrate that E2 treatment selectively shortens the fibulin-1D mRNA half-life. This indicates that estrogens affect differentially the stability of fibulin-1 variants and may explain the lower accumulation of fibulin-1D mRNA on E2 treatment. In conclusion, our data show that estrogens, via ERalpha, are key regulators of fibulin-1 expression at both the transcriptional and posttranscriptional levels. The preferential induction of the fibulin-1C variant, which is overexpressed in ovarian and breast cancer, might play an important role in estrogen-promoted carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Ovarian Neoplasms/genetics , Base Sequence , Breast Neoplasms/physiopathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Sequence Data , Ovarian Neoplasms/physiopathology , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
8.
J Mol Endocrinol ; 32(2): 583-94, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15072561

ABSTRACT

In this study, we have analysed the effects of histone deacetylase (HDAC) inhibition on estrogen receptor (ER) expression and on its transcriptional activity in response to antiestrogens. In several breast cancer cell lines, trichostatin A (TSA), a potent HDAC inhibitor, strongly decreases ERalpha expression in a dose-dependent manner. This repression is observed independently of the presence of ligand and also occurs in ovarian and endometrial cell lines. In addition, we show that in MCF7 cells bearing a stably transfected reporter plasmid (MELN cells), partial antiestrogens such as 4-OH-tamoxifen (OHTam), raloxifen or LY117018, switch to an agonist activity upon HDAC inhibition. This effect is blocked by the pure antiestrogen ICI182780 and exhibits a half-maximal concentration of OHTam equivalent to its affinity for ERalpha. The TSA-dependent decrease of ERalpha expression is required to induce the agonist switch of OHTam properties as it is lost in cells constitutively expressing exogenous receptors (MELN-ERalpha or ERbeta). By contrast, the transrepression activity of OHTam is abolished by TSA independently of the decrease of ERalpha expression. Interestingly, in MELN-ERalpha, ICI182780 remains inhibitory suggesting the involvement of HDAC-independent mechanisms. Finally, in the absence of TSA, transcriptional activity in response to OHTam is significantly raised in MELN cells expressing low levels of ERalpha after transfection of antisense oligonucleotides. In conclusion, inhibition of HDAC enzymatic activity and modulation of ERalpha levels tightly control the relative agonist activity of partial antiestrogens on a stably integrated reporter transgene.


Subject(s)
Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/metabolism , Histone Deacetylase Inhibitors , Tamoxifen/analogs & derivatives , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/drug effects , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Fulvestrant , Genes, Reporter , Humans , Hydroxamic Acids/pharmacology , Pyrrolidines/pharmacology , Raloxifene Hydrochloride/pharmacology , Response Elements/drug effects , Response Elements/genetics , Tamoxifen/pharmacology , Thiophenes/pharmacology , Transcription, Genetic , Tumor Cells, Cultured
10.
J Endocrinol ; 179(1): 41-53, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14529564

ABSTRACT

We analysed the antiproliferative activity of various histone deacetylase (HDAC) inhibitors such as trichostatin A (TSA) on human breast cancer cells. We observed a lower sensitivity to HDAC inhibition for oestrogen receptor negative (ER-) versus positive (ER+) cell lines. This differential response was associated neither with a modification of drug efflux via the multidrug resistance system nor with a global modification of histone acetyltransferase (HAT)/HDAC activities. In contrast, we demonstrated that in ER+ breast cancer cells the p21(WAF1/CIP1) gene was more sensitive to TSA regulation and was expressed at higher levels. These differences were observed both in transient transfection experiments and on the endogenous p21(WAF1/CIP1) gene. The Sp1 transcription factor, which was shown to interact in vitro with both class I and class II HDACs, is sufficient to confer the differential sensitivity to TSA and participated in the control of p21(WAF1/CIP1) basal expression. Finally, re-expression of ERalpha following adenoviral infection of ER- breast cancer cells increased both p21(WAF1/CIP1) protein accumulation and the growth inhibitory activity of TSA. Altogether, our results highlight the key role of ERalpha and p21(WAF1/CIP1) gene expression in the sensitivity of breast cancer cells to hyperacetylating agents.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cyclins/genetics , Histone Deacetylase Inhibitors , Receptors, Estrogen/physiology , Acetylation/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Estrogen Receptor alpha , Female , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Estrogen/metabolism , Sp1 Transcription Factor/physiology , Transfection , Tumor Cells, Cultured
11.
Mol Cell Biol ; 19(10): 6488-99, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490589

ABSTRACT

To study the molecular mechanisms of circadian gene expression, we have sought to identify genes whose expression in mouse liver is regulated by the transcription factor DBP (albumin D-site-binding protein). This PAR basic leucine zipper protein accumulates according to a robust circadian rhythm in nuclei of hepatocytes and other cell types. Here, we report that the Cyp2a4 gene, encoding the cytochrome P450 steroid 15alpha-hydroxylase, is a novel circadian expression gene. This enzyme catalyzes one of the hydroxylation reactions leading to further metabolism of the sex hormones testosterone and estradiol in the liver. Accumulation of CYP2A4 mRNA in mouse liver displays circadian kinetics indistinguishable from those of the highly related CYP2A5 gene. Proteins encoded by both the Cyp2a4 and Cyp2a5 genes also display daily variation in accumulation, though this is more dramatic for CYP2A4 than for CYP2A5. Biochemical evidence, including in vitro DNase I footprinting on the Cyp2a4 and Cyp2a5 promoters and cotransfection experiments with the human hepatoma cell line HepG2, suggests that the Cyp2a4 and Cyp2a5 genes are indeed regulated by DBP. These conclusions are corroborated by genetic studies, in which the circadian amplitude of CYP2A4 and CYP2A5 mRNAs and protein expression in the liver was significantly impaired in a mutant mouse strain homozygous for a dbp null allele. These experiments strongly suggest that DBP is a major factor controlling circadian expression of the Cyp2a4 and Cyp2a5 genes in the mouse liver.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Circadian Rhythm , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins , Microsomes, Liver/enzymology , Mixed Function Oxygenases/genetics , Steroid Hydroxylases/genetics , Transcription Factors/metabolism , Animals , Binding Sites , Cytochrome P-450 CYP2A6 , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P450 Family 2 , DNA Footprinting , Gene Expression Regulation, Enzymologic , Homozygote , Humans , Leucine Zippers , Mice , Mice, Knockout , Mixed Function Oxygenases/biosynthesis , Mutation , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/biosynthesis , Steroid Hydroxylases/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...