850 resultados para Histone Deacetylase Inhibition
Resumo:
Purpose: To characterize the importance of cellular Fas-associated death domain (FADD)–like interleukin 1ß-converting enzyme (FLICE) inhibitory protein (c-FLIP), a key regulator of caspase-8 (FLICE)–promoted apoptosis, in modulating the response of prostate cancer cells to androgen receptor (AR)–targeted therapy.
Experimental Design: c-FLIP expression was characterized by immunohistochemical analysis of prostatectomy tissue. The functional importance of c-FLIP to survival and modulating response to bicalutamide was studied by molecular and pharmacologic interventions.
Results: c-FLIP expression was increased in high-grade prostatic intraepithelial neoplasia and prostate cancer tissue relative to normal prostate epithelium (P < 0.001). Maximal c-FLIP expression was detected in castrate-resistant prostate cancer (CRPC; P < 0.001). In vitro, silencing of c-FLIP induced spontaneous apoptosis and increased 22Rv1 and LNCaP cell sensitivity to bicalutamide, determined by flow cytometry, PARP cleavage, and caspase activity assays. The histone deacetylase inhibitors (HDACi), droxinostat and SAHA, also downregulated c-FLIP expression, induced caspase-8- and caspase-3/7–mediated apoptosis, and increased apoptosis in bicalutamide-treated cells. Conversely, the elevated expression of c-FLIP detected in the CRPC cell line VCaP underpinned their insensitivity to bicalutamide and SAHA in vitro. However, knockdown of c-FLIP induced spontaneous apoptosis in VCaP cells, indicating its relevance to cell survival and therapeutic resistance.
Conclusion: c-FLIP reduces the efficacy of AR-targeted therapy and maintains the viability of prostate cancer cells. A combination of HDACi with androgen deprivation therapy may be effective in early-stage disease, using c-FLIP expression as a predictive biomarker of sensitivity. Direct targeting of c-FLIP, however, may be relevant to enhance the response of existing and novel therapeutics in CRPC. Clin Cancer Res; 18(14); 3822–33.
Resumo:
We have previously demonstrated that histone deacetylase 7 (HDAC7) expression and splicing play an important role in smooth muscle cell (SMC) differentiation from embryonic stem (ES) cells, but the molecular mechanisms of increased HDAC7 expression during SMC differentiation are currently unknown. In this study, we found that platelet-derived growth factor-BB (PDGF-BB) induced a 3-fold increase in the transcripts of HDAC7 in differentiating ES cells. Importantly, our data also revealed that PDGF-BB regulated HDAC7 expression not through phosphorylation of HDAC7 but through transcriptional activation. By dissecting its promoters with progressive deletion analysis, we identified the sequence between -343 and -292 bp in the 5'-flanking region of the Hdac7 gene promoter as the minimal PDGF-BB-responsive element, which contains one binding site for the transcription factor, specificity protein 1 (Sp1). Mutation of the Sp1 site within this PDGF-BB-responsive element abolished PDGF-BB-induced HDAC7 activity. PDGF-BB treatment enhanced Sp1 binding to the Hdac7 promoter in differentiated SMCs in vivo as demonstrated by the chromatin immunoprecipitation assay. Moreover, we also demonstrated that knockdown of Sp1 abrogated PDGF-BB-induced HDAC7 up-regulation and SMC differentiation gene expression in differentiating ES cells, although enforced expression of Sp1 alone was sufficient to increase the activity of the Hdac7 promoter and expression levels of SMC differentiation genes. Importantly, we further demonstrated that HDAC7 was required for Sp1-induced SMC differentiation of gene expression. Our data suggest that Sp1 plays an important role in the regulation of Hdac7 gene expression in SMC differentiation from ES cells. These findings provide novel molecular insights into the regulation of HDAC7 and enhance our knowledge in SMC differentiation and vessel formation during embryonic development.
Resumo:
Reendothelialization involves endothelial progenitor cell (EPC) homing, proliferation, and differentiation, which may be influenced by fluid shear stress and local flow pattern. This study aims to elucidate the role of laminar flow on embryonic stem (ES) cell differentiation and the underlying mechanism. We demonstrated that laminar flow enhanced ES cell-derived progenitor cell proliferation and differentiation into endothelial cells (ECs). Laminar flow stabilized and activated histone deacetylase 3 (HDAC3) through the Flk-1-PI3K-Akt pathway, which in turn deacetylated p53, leading to p21 activation. A similar signal pathway was detected in vascular endothelial growth factor-induced EC differentiation. HDAC3 and p21 were detected in blood vessels during embryogenesis. Local transfer of ES cell-derived EPC incorporated into injured femoral artery and reduced neointima formation in a mouse model. These data suggest that shear stress is a key regulator for stem cell differentiation into EC, especially in EPC differentiation, which can be used for vascular repair, and that the Flk-1-PI3K-Akt-HDAC3-p53-p21 pathway is crucial in such a process.
Resumo:
BACKGROUND: Cells deploy quality control mechanisms to remove damaged or misfolded proteins. Recently, we have reported that a mutation (R43W) in the Frank-ter Haar syndrome protein Tks4 resulted in aberrant intracellular localization.
RESULTS: Here we demonstrate that the accumulation of Tks4(R43W) depends on the intact microtubule network. Detergent-insoluble Tks4 mutant colocalizes with the centrosome and its aggregate is encaged by the intermediate filament protein vimentin. Both the microtubule inhibitor nocodazole and the histone deacetylase inhibitor Trichostatin A inhibit markedly the aggresome formation in cells expressing Tks4(R43W). Finally, pretreatment of cells with the proteasome inhibitor MG132 markedly increases the level of aggresomes formed by Tks4(R43W). Furthermore, two additional mutant Tks4 proteins (Tks4(1-48) or Tks4(1-341)) have been investigated. Whereas the shorter Tks4 mutant, Tks4(1-48), shows no expression at all, the longer Tks4 truncation mutant accumulates in the nuclei of the cells.
CONCLUSIONS: Our results suggest that misfolded Frank-ter Haar syndrome protein Tks4(R43W) is transported via the microtubule system to the aggresomes. Lack of expression of Tks4(1-48) or aberrant intracellular expressions of Tks4(R43W) and Tks4(1-341) strongly suggest that these mutations result in dysfunctional proteins which are not capable of operating properly, leading to the development of FTHS.
Resumo:
Cervical cancer is a multi-stage disease caused by human papillomaviruses (HPV) infection of cervical epithelial cells, but the mechanisms regulating disease progression are not clearly defined. Using 3-dimensional organotypic cultures, we demonstrate that HPV16 E6 and E7 proteins alter the secretome of primary human keratinocytes resulting in local epithelial invasion. Mechanistically, absence of the IGF-binding protein 2 (IGFBP2) caused increases in IGFI/II signalling and through crosstalk with KGF/FGFR2b/AKT, cell invasion. Repression of IGFBP2 is mediated by histone deacetylation at the IGFBP2 promoter and was reversed by treatment with histone deacetylase (HDAC) inhibitors. Our in vitro findings were confirmed in 50 invasive cancers and 79 cervical intra-epithelial neoplastic lesions caused by HPV16 infection, where IGFBP2 levels were reduced with increasing disease severity. In summary, the loss of IGFBP2 is associated with progression of premalignant disease, and sensitises cells to pro-invasive IGF signalling, and together with stromal derived factors promotes epithelial invasion.
Resumo:
During cancer development and progression, tumor cells undergo abnormal epigenetic modifications, including DNA methylation, histone deacetylation and nucleosome remodeling. Collectively, these aberrations promote genomic instability and lead to silencing of tumor-suppressor genes and reactivation of oncogenic retroviruses. Epigenetic modifications, therefore, provide exciting new avenues for prostate cancer research. Promoter hypermethylation is widespread during neoplastic transformation of prostate cells, which suggests that restoration of a 'normal' epigenome through treatment with inhibitors of the enzymes involved could be clinically beneficial. Global patterns of histone modifications are also being defined and have been associated with clinical and pathologic predictors of prostate cancer outcome. Although treatment for localized prostate cancer can be curative, the development of successful therapies for the management of castration-resistant metastatic disease is urgently needed. Reactivation of tumor-suppressor genes by demethylating agents and histone deacetylase inhibitors could be a potential treatment option for patients with advanced disease.
Resumo:
Higher expression of the miR-433 microRNA (miRNA) is associated with poorer survival outcomes in patients with HGSOC that may be overcome by a greater understanding of the functional role of this miRNA. We previously described miR-433 as a critical cell cycle regulator and mediator of cellular senescence. Downregulation of the mitotic arrest deficiency 2 (MAD2) protein by miR-433 led to increased cellular resistance to paclitaxel in epithelial ovarian cancer cells (EOC). Furthermore immunohistochemical (IHC) analysis of MAD2 expression in patients with HGSOC showed that loss of MAD2 was significantly associated with poorer patient survival. Higher miR-433 expression is also associated with an increased resistance to the platins which is unrelated to loss of MAD2 expression. In silico analysis of the miR-433 target proteins in the TCGA database identified the association between a number of miR-433 targets and poorer patient survival. IHC analysis of the miR-433 target, histone deacetylase 6 (HDAC6), confirmed that its expression was significantly associated with a decrease in patient overall survival. The knock-down of HDAC6 by siRNA in EOC cells did not attenuate apoptotic responses to paclitaxel or platin although lower endogenous HDAC6 expression was associated with more resistant EOC cell lines. In vitro analysis revealed that EOC cells which survived chemotherapeutic kill with high doses of paclitaxel expressed higher miR-433 and concomitant decreased expression of the miR-433 targets. These cells were more chemoresistant compared to the parental cell line and repopulated as 3d organoid cultures in non-adherent stem cell selective conditions; thus indicating that the cells which survive chemotherapy were viable, capable of regrowth and had an increased potential for pluripotency. In conclusion, our data suggests that chemotherapy is not driving the transcriptional upregulation of miR-433 but rather selecting a population of cells with high miR-433 expression that may contribute to chemoresistant disease and tumour recurrence.
Resumo:
Increasingly invasive bladder cancer cells lines displayed insensitivity toward a panel of dietary-derived ligands for members of the nuclear receptor superfamily. Insensitivity was defined through altered gene regulatory actions and cell proliferation and reflected both reduced receptor expression and elevated nuclear receptor corepressor 1 (NCOR1) expression. Stable overexpression of NCOR1 in sensitive cells (RT4) resulted in a panel of clones that recapitulated the resistant phenotype in terms of gene regulatory actions and proliferative responses toward ligand. Similarly, silencing RNA approaches to NCOR1 in resistant cells (EJ28) enhanced ligand gene regulatory and proliferation responses, including those mediated by peroxisome proliferator-activated receptor (PPAR) gamma and vitamin D receptor (VDR) receptors. Elevated NCOR1 levels generate an epigenetic lesion to target in resistant cells using the histone deacetylase inhibitor vorinostat, in combination with nuclear receptor ligands. Such treatments revealed strong-additive interactions toward the PPARgamma, VDR and Farnesoid X-activated receptors. Genome-wide microarray and microfluidic quantitative real-time, reverse transcription-polymerase chain reaction approaches, following the targeting of NCOR1 activity and expression, revealed the selective capacity of this corepressor to govern common transcriptional events of underlying networks. Combined these findings suggest that NCOR1 is a selective regulator of nuclear receptors, notably PPARgamma and VDR, and contributes to their loss of sensitivity. Combinations of epigenetic therapies that target NCOR1 may prove effective, even when receptor expression is reduced.
Resumo:
Methamphetamine (METH) is a powerful psychostimulant drug used worldwide for its reinforcing properties. In addition to the classic long-lasting monoaminergic-disrupting effects extensively described in the literature, METH has been consistently reported to increase blood brain barrier (BBB) permeability, both in vivo and in vitro, as a result of tight junction and cytoskeleton disarrangement. Microtubules play a critical role in cell stability, which relies on post-translational modifications such as a-tubulin acetylation. As there is evidence that psychostimulants drugs modulate the expression of histone deacetylases (HDACs), we hypothesized that in endothelial cells METH-mediation of cytoplasmatic HDAC6 activity could affect tubulin acetylation and further contribute to BBB dysfunction. To validate our hypothesis, we exposed the bEnd.3 endothelial cells to increasing doses of METH and verified that itleads to an extensivea-tubulin deacetylation mediated by HDACs activation. Furthermore, since we recently reported that acetyl-L-carnitine (ALC), a natural occurring compound, prevents BBB structural loss in a context of METH exposure, we reasoned that ALC could also preserve the acetylation of microtubules under METH action. The present results confirm that ALC is able to prevent METH-induced deacetylation providing effective protection on microtubule acetylation. Although further investigation is still needed, HDACs regulation may become a new therapeutic target for ALC.
Resumo:
The need for better gene transfer systems towards improved risk=benefit balance for patients remains a major challenge in the clinical translation of gene therapy (GT). We have investigated the improvement of integrating vectors safety in combining (i) new short synthetic genetic insulator elements (GIE) and (ii) directing genetic integration to heterochromatin. We have designed SIN-insulated retrovectors with two candidate GIEs and could identify a specific combination of insulator 2 repeats which translates into best functional activity, high titers and boundary effect in both gammaretro (p20) and lentivectors (DCaro4) (see Duros et al, abstract ibid). Since GIEs are believed to shield the transgenic cassette from inhibitory effects and silencing, DCaro4 has been further tested with chimeric HIV-1 derived integrases which comprise C-ter chromodomains targeting heterochromatin through either histone H3 (ML6chimera) or methylatedCpGislands (ML10). With DCaro4 only and both chimeras, a homogeneous expression is evidenced in over 20% of the cells which is sustained over time. With control lentivectors, less than 2% of cells express GFP as compared to background using a control double-mutant in both catalytic and ledgf binding-sites; in addition, a two-times increase of expression can be induced with histone deacetylase inhibitors. Our approach could significantly reduce integration into open chromatin sensitive sites in stem cells at the time of transduction, a feature which might significantly decrease subsequent genotoxicity, according to X-SCIDs patients data.Work performed with the support of EC-DG research within the FP6-Network of Excellence, CLINIGENE: LSHB-CT-2006-018933
Resumo:
La chromatine est un système de compaction de l’ADN jouant un rôle important dans la régulation de l’expression génique. L’acétylation de la chromatine provoque un relâchement de sa structure, facilitant le recrutement de facteurs de transcription. Inversement, des complexes histones déacétylases favorisent une structure compacte, réprimant l’expression de gènes. Un complexe HDAC, Rpd3S, est recruté par l’ARN polymérase II phosphorylée sur les régions codantes transcrites. Cette activité HDAC est stimulée par la déposition de la marque H3K36me générée par l’histone méthyltransférase Set2. Par approche génomique, en utilisant comme organisme modèle Saccharomyces cerevisiae, j’ai optimisé la méthode de ChIP-chip puis démontré que les sous unités Hos2 et Set3 d’un autre complexe HDAC, Set3C, étaient recrutées sur des régions codantes de gènes transcrits. De plus, Set3C est connu pour être recruté soit par H3K4me ou par la Pol II. La suite du projet portera sur le recrutement de Set3C qui semble similaire à Rpd3S.
Resumo:
Au cours des maladies cardiovasculaires (MCV), il peut se produire divers problèmes de santé, telle que l’insuffisance cardiaque ou encore l’HTA. Ces phénomènes se caractérisent, entre autres, par une augmentation de synthèse d’endotheline-1 (ET-1), un neuropeptide synthétisé par les cellules endothéliales ayant un effet vasoconstricteur sur les cellules musculaires lisses vasculaires (CMLV). Ainsi, la surexpression de ce vasopeptide, mène à terme, au maintien de l’HTA aggravée des sujets, précédée ou concomitante à l’athérosclérose ou à la resténose, cliniquement illustrées par une prolifération et une migration anormale des CMLV de la media vers l’intima des vaisseaux sanguins. Parallèlement, il a été observé que la protéine sirtuine-1 (Sirt-1), membre de la famille des protéines histones déacétylases (HDAC), présente des propriétés anti-athérosclérotiques par sa capacité d’atténuer la prolifération et la migration des CMLV. Des travaux récents ont aussi montré qu’au cours de l’HTA la protéine Sirt-1 est faiblement exprimée dans les CMLV. Son implication dans le développement des pathologies vasculaires semble apparente, mais des études demeurent nécessaires pour décrire son rôle exact dans la pathogenèse des MCV. Dans cette optique, l’objectif de cette étude a été d’observer la variation d’expression de Sirt-1 dans les CMLV, isolées de l’aorte ascendante de rat, en réponse à l’ET-1. On a remarqué qu’une heure de stimulation des CMLV avec l’ET-1 induit une diminution de l’expression de Sirt-1 via l’activation des récepteurs ETA. Ces résultats suggèrent que la capacité d’ET-1 à atténuer l’expression de Sirt-1 serait un éventuel mécanisme d’action avec des effets favorisant les MCV.
Resumo:
La sénescence cellulaire, ou l’arrêt irréversible de la prolifération, influence des processus physiologiques et pathologiques, comme le cancer. Parmi les caractéristiques de la sénescence, se retrouve le PSAS ou phénotype sécrétoire associé à la sénescence. Le PSAS est composé d’une variété de cytokines, facteurs de croissance et protéases. Ses actions pro- et anti-tumorale sont connues, mais l’on ignore laquelle prédomine. Mes travaux s’attardent aux effets du PSAS sur l’induction de la sénescence dans les cellules environnantes et à sa régulation. Nous avons démontré que le PSAS ne synergise pas avec la dysfonction télomérique chronique ou aigue, afin de causer un arrêt de croissance. Également, l’étude du mécanisme responsable de l’induction de la sénescence par stress à la chromatine, suggère que la kinase c-Abl n’est pas requise pour cette voie, contrairement à des publications antérieures. Mes travaux éclairent les mécanismes d’action et la régulation du PSAS dans la sénescence induite par dysfonction télomérique et par stress à la chromatine.
Resumo:
La chromatine possède une plasticité complexe et essentielle pour répondre à différents mécanismes cellulaires fondamentaux tels la réplication, la transcription et la réparation de l’ADN. Les histones sont les constituants essentiels de la formation des nucléosomes qui assurent le bon fonctionnement cellulaire d’où l’intérêt de cette thèse d’y porter une attention particulière. Un dysfonctionnement de la chromatine est souvent associé à l’émergence du cancer. Le chapitre II de cette thèse focalise sur la répression transcriptionnelle des gènes d’histones par le complexe HIR (HIstone gene Repressor) en réponse au dommage à l'ADN chez Saccharomyces cerevisiae. Lors de dommage à l’ADN en début de phase S, les kinases du point de contrôle Mec1, Tel1 et Rad53 s’assurent de bloquer les origines tardives de réplication pour limiter le nombre de collisions potentiellement mutagéniques ou cytotoxiques entre les ADN polymérases et les lésions persistantes dans l'ADN. Lorsque la synthèse totale d’ADN est soudainement ralentie par le point de contrôle, l’accumulation d'un excès d'histones nouvellement synthétisées est néfaste pour les cellules car les histones libres se lient de manière non-spécifique aux acides nucléiques. L'un des mécanismes mis en place afin de minimiser la quantité d’histones libres consiste à réprimer la transcription des gènes d'histones lors d'une chute rapide de la synthèse d'ADN, mais les bases moléculaires de ce mécanisme étaient très mal connues. Notre étude sur la répression des gènes d’histones en réponse aux agents génotoxiques nous a permis d’identifier que les kinases du point de contrôle jouent un rôle dans la répression des gènes d’histones. Avant le début de mon projet, il était déjà connu que le complexe HIR est requis pour la répression des gènes d’histones en phase G1, G2/M et lors de dommage à l’ADN en phase S. Par contre, la régulation du complexe HIR en réponse au dommage à l'ADN n'était pas connue. Nous avons démontré par des essais de spectrométrie de masse (SM) que Rad53 régule le complexe HIR en phosphorylant directement une de ses sous-unités, Hpc2, à de multiples résidus in vivo et in vitro. La phosphorylation d’Hpc2 est essentielle pour le recrutement aux promoteurs de gènes d’histones du complexe RSC (Remodels the Structure of Chromatin) dont la présence sur les promoteurs des gènes d'histones corrèle avec leur répression. De plus, nous avons mis à jour un nouveau mécanisme de régulation du complexe HIR durant la progression normale à travers le cycle cellulaire ainsi qu'en réponse aux agents génotoxiques. En effet, durant le cycle cellulaire normal, la protéine Hpc2 est très instable durant la transition G1/S afin de permettre la transcription des gènes d’histones et la production d'un pool d'histones néo-synthétisées juste avant l'initiation de la réplication de l’ADN. Toutefois, Hpc2 n'est instable que pour une brève période de temps durant la phase S. Ces résultats suggèrent qu'Hpc2 est une protéine clef pour la régulation de l'activité du complexe HIR et la répression des gènes d’histones lors du cycle cellulaire normal ainsi qu'en réponse au dommage à l’ADN. Dans le but de poursuivre notre étude sur la régulation des histones, le chapitre III de ma thèse concerne l’analyse globale de l’acétylation des histones induite par les inhibiteurs d’histone désacétylases (HDACi) dans les cellules normales et cancéreuses. Les histones désacétylases (HDACs) sont les enzymes qui enlèvent l’acétylation sur les lysines des histones. Dans plusieurs types de cancers, les HDACs contribuent à l’oncogenèse par leur fusion aberrante avec des complexes protéiques oncogéniques. Les perturbations causées mènent souvent à un état silencieux anormal des suppresseurs de tumeurs. Les HDACs sont donc une cible de choix dans le traitement des cancers engendrés par ces protéines de fusion. Notre étude de l’effet sur l’acétylation des histones de deux inhibiteurs d'HDACs de relevance clinique, le vorinostat (SAHA) et l’entinostat (MS-275), a permis de démontrer une augmentation élevée de l’acétylation globale des histones H3 et H4, contrairement à H2A et H2B, et ce, autant chez les cellules normales que cancéreuses. Notre quantification en SM de l'acétylation des histones a révélé de façon inattendue que la stœchiométrie d'acétylation sur la lysine 56 de l’histone H3 (H3K56Ac) est de seulement 0,03% et, de manière surprenante, cette stœchiométrie n'augmente pas dans des cellules traitées avec différents HDACi. Plusieurs études de H3K56Ac chez l’humain présentes dans la littérature ont rapporté des résultats irréconciliables. Qui plus est, H3K56Ac était considéré comme un biomarqueur potentiel dans le diagnostic et pronostic de plusieurs types de cancers. C’est pourquoi nous avons porté notre attention sur la spécificité des anticorps utilisés et avons déterminé qu’une grande majorité d’anticorps utilisés dans la littérature reconnaissent d’autres sites d'acétylation de l’histone H3, notamment H3K9Ac dont la stœchiométrie d'acétylation in vivo est beaucoup plus élevée que celle d'H3K56Ac. De plus, le chapitre IV fait suite à notre étude sur l’acétylation des histones et consiste en un rapport spécial de recherche décrivant la fonction de H3K56Ac chez la levure et l’homme et comporte également une évaluation d’un anticorps supposément spécifique d'H3K56Ac en tant qu'outil diagnostic du cancer chez l’humain.
Resumo:
Histone deacetylase inhibitors (HDACIs) interfere with the epigenetic process of histone acetylation and are known to have analgesic properties in models of chronic inflammatory pain. The aim of this study was to determine whether these compounds could also affect neuropathic pain. Different class I HDACIs were delivered intrathecally into rat spinal cord in models of traumatic nerve injury and antiretroviral drug-induced peripheral neuropathy (stavudine, d4T). Mechanical and thermal hypersensitivity was attenuated by 40% to 50% as a result of HDACI treatment, but only if started before any insult. The drugs globally increased histone acetylation in the spinal cord, but appeared to have no measurable effects in relevant dorsal root ganglia in this treatment paradigm, suggesting that any potential mechanism should be sought in the central nervous system. Microarray analysis of dorsal cord RNA revealed the signature of the specific compound used (MS-275) and suggested that its main effect was mediated through HDAC1. Taken together, these data support a role for histone acetylation in the emergence of neuropathic pain.