Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
2.
Mol Cancer Ther ; 22(1): 52-62, 2023 01 03.
Article de Anglais | MEDLINE | ID: mdl-36343387

RÉSUMÉ

The EGFR/RAS/MEK/ERK signaling pathway (ERK/MAPK) is hyperactivated in most colorectal cancers. A current limitation of inhibitors of this pathway is that they primarily induce cytostatic effects in colorectal cancer cells. Nevertheless, these drugs do induce expression of proapoptotic factors, suggesting they may prime colorectal cancer cells to undergo apoptosis. As histone deacetylase inhibitors (HDACis) induce expression of multiple proapoptotic proteins, we examined whether they could synergize with ERK/MAPK inhibitors to trigger colorectal cancer cell apoptosis. Combined MEK/ERK and HDAC inhibition synergistically induced apoptosis in colorectal cancer cell lines and patient-derived tumor organoids in vitro, and attenuated Apc-initiated adenoma formation in vivo. Mechanistically, combined MAPK/HDAC inhibition enhanced expression of the BH3-only proapoptotic proteins BIM and BMF, and their knockdown significantly attenuated MAPK/HDAC inhibitor-induced apoptosis. Importantly, we demonstrate that the paradigm of combined MAPK/HDAC inhibitor treatment to induce apoptosis can be tailored to specific MAPK genotypes in colorectal cancers, by combining an HDAC inhibitor with either an EGFR, KRASG12C or BRAFV600 inhibitor in KRAS/BRAFWT; KRASG12C, BRAFV600E colorectal cancer cell lines, respectively. These findings identify a series of ERK/MAPK genotype-tailored treatment strategies that can readily undergo clinical testing for the treatment of colorectal cancer.


Sujet(s)
Tumeurs colorectales , Inhibiteurs de désacétylase d'histone , Humains , Apoptose , Protéines régulatrices de l'apoptose , Mort cellulaire , Lignée cellulaire tumorale , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Récepteurs ErbB , Inhibiteurs de désacétylase d'histone/pharmacologie , Mitogen-Activated Protein Kinase Kinases , Système de signalisation des MAP kinases
3.
Cell Death Differ ; 29(11): 2288-2302, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35606410

RÉSUMÉ

Colorectal cancers (CRCs) often display histological features indicative of aberrant differentiation but the molecular underpinnings of this trait and whether it directly drives disease progression is unclear. Here, we identify co-ordinate epigenetic inactivation of two epithelial-specific transcription factors, EHF and CDX1, as a mechanism driving differentiation loss in CRCs. Re-expression of EHF and CDX1 in poorly-differentiated CRC cells induced extensive chromatin remodelling, transcriptional re-programming, and differentiation along the enterocytic lineage, leading to reduced growth and metastasis. Strikingly, EHF and CDX1 were also able to reprogramme non-colonic epithelial cells to express colonic differentiation markers. By contrast, inactivation of EHF and CDX1 in well-differentiated CRC cells triggered tumour de-differentiation. Mechanistically, we demonstrate that EHF physically interacts with CDX1 via its PNT domain, and that these transcription factors co-operatively drive transcription of the colonic differentiation marker, VIL1. Compound genetic deletion of Ehf and Cdx1 in the mouse colon disrupted normal colonic differentiation and significantly enhanced colorectal tumour progression. These findings thus reveal a novel mechanism driving epithelial de-differentiation and tumour progression in CRC.


Sujet(s)
Tumeurs colorectales , Facteurs de transcription , Animaux , Souris , Tumeurs colorectales/génétique , Épigenèse génétique , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
4.
Trends Cancer ; 3(10): 698-712, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28958388

RÉSUMÉ

Colorectal cancers (CRCs) are characterized by global hypomethylation and promoter-specific DNA methylation. A subset of CRCs with extensive and co-ordinate patterns of promoter methylation has also been identified, termed the CpG-island methylator phenotype. Some genes methylated in CRC are established tumor suppressors; however, for the majority, direct roles in disease initiation or progression have not been established. Herein, we examine functional evidence of specific methylated genes contributing to CRC pathogenesis, focusing on components of commonly deregulated signaling pathways. We also review current knowledge of the mechanisms underpinning promoter methylation in CRC, including genetic events, altered transcription factor binding, and DNA damage. Finally, we summarize clinical trials of DNA methyltransferase inhibitors in CRC, and propose strategies for enhancing their efficacy.


Sujet(s)
Tumeurs colorectales/génétique , Méthylation de l'ADN , Épigenèse génétique , Animaux , Marqueurs biologiques tumoraux , Essais cliniques comme sujet , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Association thérapeutique , DNA (cytosine-5-)-methyltransferase/antagonistes et inhibiteurs , DNA (cytosine-5-)-methyltransferase/métabolisme , Altération de l'ADN , Réparation de l'ADN , Régulation de l'expression des gènes tumoraux , Étude d'association pangénomique , Humains , Thérapie moléculaire ciblée , Régions promotrices (génétique) , Séquences répétées d'acides nucléiques , Transduction du signal
5.
Clin Cancer Res ; 23(18): 5573-5584, 2017 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-28611196

RÉSUMÉ

Purpose: Histone deacetylase inhibitors (HDACi) are epigenome-targeting small molecules approved for the treatment of cutaneous T-cell lymphoma and multiple myeloma. They have also demonstrated clinical activity in acute myelogenous leukemia, non-small cell lung cancer, and estrogen receptor-positive breast cancer, and trials are underway assessing their activity in combination regimens including immunotherapy. However, there is currently no clear strategy to reliably predict HDACi sensitivity. In colon cancer cells, apoptotic sensitivity to HDACi is associated with transcriptional induction of multiple immediate-early (IE) genes. Here, we examined whether this transcriptional response predicts HDACi sensitivity across tumor type and investigated the mechanism by which it triggers apoptosis.Experimental Design: Fifty cancer cell lines from diverse tumor types were screened to establish the correlation between apoptotic sensitivity, induction of IE genes, and components of the intrinsic apoptotic pathway.Results: We show that sensitivity to HDACi across tumor types is predicted by induction of the IE genes FOS, JUN, and ATF3, but that only ATF3 is required for HDACi-induced apoptosis. We further demonstrate that the proapoptotic function of ATF3 is mediated through direct transcriptional repression of the prosurvival factor BCL-XL (BCL2L1) These findings provided the rationale for dual inhibition of HDAC and BCL-XL, which we show strongly cooperate to overcome inherent resistance to HDACi across diverse tumor cell types.Conclusions: These findings explain the heterogeneous responses of tumor cells to HDACi-induced apoptosis and suggest a framework for predicting response and expanding their therapeutic use in multiple cancer types. Clin Cancer Res; 23(18); 5573-84. ©2017 AACR.


Sujet(s)
Facteur de transcription ATF-3/métabolisme , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Tumeurs/métabolisme , Protéine bcl-X/métabolisme , Facteur de transcription ATF-3/génétique , Animaux , Apoptose/génétique , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques , Expression des gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Gènes précoces , Gènes rapporteurs , Humains , Souris , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Interférence par ARN , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Protéine bcl-X/génétique
6.
Mol Oncol ; 11(9): 1130-1142, 2017 09.
Article de Anglais | MEDLINE | ID: mdl-28544747

RÉSUMÉ

Advanced biliary tract cancer (BTC) has a poor prognosis and limited treatment options. The PI3K/Akt/mTOR signalling pathway is hyperactivated in a subset of BTCs, and clinical activity of the mTOR inhibitor everolimus has been observed in some patients with BTC. The goal of this study was to identify biomarkers predictive of everolimus response. Twenty BTC cell lines were assessed for everolimus sensitivity with a spectrum of growth inhibitory responses observed. Molecular biomarkers of sensitivity and resistance were identified by interrogation of the activation status of the Ras/MAPK and PI3K/Akt/mTOR pathways. K-Ras mutations and/or amplifications were identified in 45% of cell lines and were associated with resistance to everolimus. Activating mutations in PIK3CA or loss of PTEN was not predictive of everolimus response; however, high basal levels of pAKT were associated with sensitivity, independent of Ras/MAPK pathway activation status. Notably, everolimus inhibited mTOR signalling to a similar extent in sensitive and resistant cell lines, suggesting that relative dependence on the mTOR pathway rather than the magnitude of pathway inhibition determines everolimus response. Consistent with the known limitations of rapalogs, everolimus induced feedback-mediated activation of AKT in BTC cell lines, which could be overcome by cotreatment with an AKT inhibitor or ATP-competitive mTORC1/mTORC2 inhibitors. However, both approaches failed to induce greater apoptosis compared to everolimus, and mTORC1/mTORC2 kinase inhibitors induced compensatory activation of pERK, identifying an inherent limitation of these agents in BTC cell lines. These findings suggest that future trials of everolimus in BTC would benefit from preselecting patients based on their K-Ras and PI3K/mTOR pathway activation status. The study also identifies strategies for enhancing inhibition of the PI3K/mTOR pathway in BTC cell lines.


Sujet(s)
Tumeurs des voies biliaires/traitement médicamenteux , Tumeurs des voies biliaires/génétique , Résistance aux médicaments antinéoplasiques/génétique , Évérolimus/usage thérapeutique , Amplification de gène , Mutation/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Tumeurs des voies biliaires/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Évérolimus/pharmacologie , Dosage génique , Composés hétérocycliques 3 noyaux/pharmacologie , Composés hétérocycliques 3 noyaux/usage thérapeutique , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Phosphohydrolase PTEN/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme
7.
Antioxid Redox Signal ; 23(1): 66-84, 2015 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-24512308

RÉSUMÉ

SIGNIFICANCE: Class I and II histone deacetylase inhibitors (HDACis) are approved for the treatment of cutaneous T-cell lymphoma and are undergoing clinical trials as single agents, and in combination, for other hematological and solid tumors. Understanding their mechanisms of action is essential for their more effective clinical use, and broadening their clinical potential. RECENT ADVANCES: HDACi induce extensive transcriptional changes in tumor cells by activating and repressing similar numbers of genes. These transcriptional changes mediate, at least in part, HDACi-mediated growth inhibition, apoptosis, and differentiation. Here, we highlight two fundamental mechanisms by which HDACi regulate gene expression­histone and transcription factor acetylation. We also review the transcriptional responses invoked by HDACi, and compare these effects within and across tumor types. CRITICAL ISSUES: The mechanistic basis for how HDACi activate, and in particular repress gene expression, is not well understood. In addition, whether subsets of genes are reproducibly regulated by these agents both within and across tumor types has not been systematically addressed. A detailed understanding of the transcriptional changes elicited by HDACi in various tumor types, and the mechanistic basis for these effects, may provide insights into the specificity of these drugs for transformed cells and specific tumor types. FUTURE DIRECTIONS: Understanding the mechanisms by which HDACi regulate gene expression and an appreciation of their transcriptional targets could facilitate the ongoing clinical development of these emerging therapeutics. In particular, this knowledge could inform the design of rational drug combinations involving HDACi, and facilitate the identification of mechanism-based biomarkers of response.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/métabolisme , Acétylation , Animaux , Lignée cellulaire tumorale , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...