Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Trends Mol Med ; 29(4): 297-314, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36828712

RESUMEN

Approximately 10% of all pathological mutations are nonsense mutations that are responsible for several severe genetic diseases for which no treatment regimens are currently available. The most widespread strategy for treating nonsense mutations is by enhancing ribosomal readthrough of premature termination codons (PTCs) to restore the production of the full-length protein. In the past decade several compounds with readthrough potential have been identified. However, although preclinical results on these compounds are promising, clinical studies have not yielded positive outcomes. We review preclinical and clinical research related to readthrough compounds and characterize factors that contribute to the observed translational gap.


Asunto(s)
Codón sin Sentido , Ribosomas , Humanos , Mutación , Ribosomas/genética
2.
Gastroenterology ; 161(1): 239-254.e9, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33819486

RESUMEN

BACKGROUND & AIMS: In homeostasis, intestinal cell fate is controlled by balanced gradients of morphogen signaling. The bone morphogenetic protein (BMP) pathway has a physiological, prodifferentiation role, predominantly inferred through previous experimental pathway inactivation. Intestinal regeneration is underpinned by dedifferentiation and cell plasticity, but the signaling pathways that regulate this adaptive reprogramming are not well understood. We assessed the BMP signaling landscape and investigated the impact and therapeutic potential of pathway manipulation in homeostasis and regeneration. METHODS: A novel mouse model was generated to assess the effect of the autocrine Bmp4 ligand on individual secretory cell fate. We spatiotemporally mapped BMP signaling in mouse and human regenerating intestine. Transgenic models were used to explore the functional impact of pathway manipulation on stem cell fate and intestinal regeneration. RESULTS: In homeostasis, ligand exposure reduced proliferation, expedited terminal differentiation, abrogated secretory cell survival, and prevented dedifferentiation. After ulceration, physiological attenuation of BMP signaling arose through upregulation of the secreted antagonist Grem1 from topographically distinct populations of fibroblasts. Concomitant expression supported functional compensation after Grem1 deletion from tissue-resident cells. BMP pathway manipulation showed that antagonist-mediated BMP attenuation was obligatory but functionally submaximal, because regeneration was impaired or enhanced by epithelial overexpression of Bmp4 or Grem1, respectively. Mechanistically, Bmp4 abrogated regenerative stem cell reprogramming despite a convergent impact of YAP/TAZ on cell fate in remodeled wounds. CONCLUSIONS: BMP signaling prevents epithelial dedifferentiation, and pathway attenuation through stromal Grem1 upregulation was required for adaptive reprogramming in intestinal regeneration. This intercompartmental antagonism was functionally submaximal, raising the possibility of therapeutic pathway manipulation in inflammatory bowel disease.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Colitis/metabolismo , Colon/metabolismo , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Regeneración , Animales , Comunicación Autocrina , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular , Proliferación Celular , Colitis/genética , Colitis/patología , Colon/patología , Células Epiteliales/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Mucosa Intestinal/patología , Intestino Delgado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/patología , Repitelización , Transducción de Señal
3.
Gut ; 69(6): 1092-1103, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31563876

RESUMEN

OBJECTIVE: Pathological Wnt pathway activation is a conserved hallmark of colorectal cancer. Wnt-activating mutations can be divided into: i) ligand-independent (LI) alterations in intracellular signal transduction proteins (Adenomatous polyposis coli, ß-catenin), causing constitutive pathway activation and ii) ligand-dependent (LD) mutations affecting the synergistic R-Spondin axis (RNF43, RSPO-fusions) acting through amplification of endogenous Wnt signal transmembrane transduction. Our aim was to exploit differential Wnt target gene expression to generate a mutation-agnostic biomarker for LD tumours. DESIGN: We undertook harmonised multi-omic analysis of discovery (n=684) and validation cohorts (n=578) of colorectal tumours collated from publicly available data and the Stratification in Colorectal Cancer Consortium. We used mutation data to establish molecular ground truth and subdivide lesions into LI/LD tumour subsets. We contrasted transcriptional, methylation, morphological and clinical characteristics between groups. RESULTS: Wnt disrupting mutations were mutually exclusive. Desmoplastic stromal upregulation of RSPO may compensate for absence of epithelial mutation in a subset of stromal-rich tumours. Key Wnt negative regulator genes were differentially expressed between LD/LI tumours, with targeted hypermethylation of some genes (AXIN2, NKD1) occurring even in CIMP-negative LD cancers. AXIN2 mRNA expression was used as a discriminatory molecular biomarker to distinguish LD/LI tumours (area under the curve >0.93). CONCLUSIONS: Epigenetic suppression of appropriate Wnt negative feedback loops is selectively advantageous in LD tumours and differential AXIN2 expression in LD/LI lesions can be exploited as a molecular biomarker. Distinguishing between LD/LI tumour types is important; patients with LD tumours retain sensitivity to Wnt ligand inhibition and may be stratified at diagnosis to clinical trials of Porcupine inhibitors.


Asunto(s)
Neoplasias Colorrectales/diagnóstico , Transducción de Señal/genética , Proteína Wnt1/metabolismo , Anciano , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Marcadores Genéticos/genética , Humanos , Masculino , Persona de Mediana Edad , Proteína Wnt1/genética
4.
Stem Cells ; 35(1): 147-157, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27350605

RESUMEN

Since the discovery of induced pluripotent stem cells there has been intense interest in understanding the mechanisms that allow a somatic cell to be reprogrammed back to a pluripotent state. Several groups have studied the alterations in gene expression that occur as somatic cells modify their genome to that of an embryonic stem cell. Underpinning many of the gene expression changes are modifications to the epigenetic profile of the associated chromatin. We have used a large-scale shRNA screen to identify epigenetic modifiers that act as barriers to reprogramming. We have uncovered an important role for TRIM28 in cells resisting transition between somatic and pluripotent states. TRIM28 achieves this by maintaining the H3K9me3 repressed state and keeping endogenous retroviruses (ERVs) silenced. We propose that knockdown of TRIM28 during reprogramming results in more plastic H3K9me3 domains, dysregulation of genes nearby H3K9me3 marks, and up regulation of ERVs, thus facilitating the transition through reprogramming. Stem Cells 2017;35:147-157.


Asunto(s)
Reprogramación Celular , Epigénesis Genética , Células Madre Pluripotentes/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Animales , Proliferación Celular , Reprogramación Celular/genética , Cromatina/metabolismo , Retrovirus Endógenos/metabolismo , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones Transgénicos , Modelos Biológicos , Células Madre Pluripotentes/citología , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba/genética
5.
Transgenic Res ; 26(2): 187-196, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27807665

RESUMEN

The Polycomb Group protein EZH2 is upregulated in most prostate cancers, and its overexpression is associated with poor prognosis. Most insights into the functional role of EZH2 in prostate cancer have been gained using cell lines and EZH2 inactivation studies. However, the question remains whether overexpression of EZH2 can initiate prostate tumourigenesis or drive tumour progression. Appropriate transgenic mouse models that are required to answer such questions are lacking. We developed one such transgenic mouse model for conditional overexpression of Ezh2. In this transgene, Ezh2 and Luciferase are transcribed from a single open reading frame. The latter gene enables intravital bioluminescent imaging of tissues expressing this transgene, allowing the detection of tumour outgrowth and potential metastatic progression over time. Prostate-specific Ezh2 overexpression by crossbreeding with Probasin-Cre mice led to neoplastic prostate lesions at low incidence and with a long latency. Compounding a previously described Bmi1-transgene and Pten-deficiency prostate cancer mouse model with the Ezh2 transgene did not enhance tumour progression or drive metastasis formation. In conclusion, we here report the generation of a wildtype Ezh2 overexpression mouse model that allows for intravital surveillance of tissues with activated transgene. This model will be an invaluable tool for further unravelling the role of EZH2 in cancer.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/biosíntesis , Fosfohidrolasa PTEN/genética , Complejo Represivo Polycomb 1/genética , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas/genética , Animales , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/genética , Regulación de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Neoplasias de la Próstata/patología
6.
Gut ; 66(6): 1095-1105, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27511199

RESUMEN

OBJECTIVE: The gross majority of colorectal cancer cases results from aberrant Wnt/ß-catenin signalling through adenomatous polyposis coli (APC) or CTNNB1 mutations. However, a subset of human colon tumours harbour, mutually exclusive with APC and CTNNB1 mutations, gene fusions in RSPO2 or RSPO3, leading to enhanced expression of these R-spondin genes. This suggested that RSPO activation can substitute for the most common mutations as an alternative driver for intestinal cancer. Involvement of RSPO3 in tumour growth was recently shown in RSPO3-fusion-positive xenograft models. The current study determines the extent into which solely a gain in RSPO3 actually functions as a driver of intestinal cancer in a direct, causal fashion, and addresses the in vivo activities of RSPO3 in parallel. DESIGN: We generated a conditional Rspo3 transgenic mouse model in which the Rspo3 transgene is expressed upon Cre activity. Cre is provided by cross-breeding with Lgr5-GFP-CreERT2 mice. RESULTS: Upon in vivo Rspo3 expression, mice rapidly developed extensive hyperplastic, adenomatous and adenocarcinomatous lesions throughout the intestine. RSPO3 induced the expansion of Lgr5+ stem cells, Paneth cells, non-Paneth cell label-retaining cells and Lgr4+ cells, thus promoting both intestinal stem cell and niche compartments. Wnt/ß-catenin signalling was modestly increased upon Rspo3 expression and mutant Kras synergised with Rspo3 in hyperplastic growth. CONCLUSIONS: We provide in vivo evidence that RSPO3 stimulates the crypt stem cell and niche compartments and drives rapid intestinal tumorigenesis. This establishes RSPO3 as a potent driver of intestinal cancer and proposes RSPO3 as a candidate target for therapy in patients with colorectal cancer harbouring RSPO3 fusions.


Asunto(s)
Adenocarcinoma/genética , Adenoma/genética , Carcinogénesis/genética , Neoplasias Intestinales/genética , Intestinos/patología , Células de Paneth/patología , Células Madre/patología , Trombospondinas/genética , Trombospondinas/metabolismo , Adenocarcinoma/patología , Adenoma/patología , Animales , Aumento de la Célula , Movimiento Celular/genética , Proliferación Celular/genética , Expresión Génica , Hiperplasia/genética , Hiperplasia/patología , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/patología , Ratones , Ratones Transgénicos , Mutación , Organoides/patología , Células de Paneth/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/análisis , Células Madre/química , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
7.
Oncotarget ; 7(43): 69816-69828, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27634879

RESUMEN

EZH2 inhibitors have gained great interest for their use as anti-cancer therapeutics. However, most research has focused on EZH2 mutant cancers and recently adverse effects of EZH2 inactivation have come to light. To determine whether colorectal cancer cells respond to EZH2 inhibition and to explore which factors influence the degree of response, we treated a panel of 20 organoid lines derived from human colon tumors with different concentrations of the EZH2 inhibitor GSK126. The resulting responses were associated with mutation status, gene expression and responses to other drugs. We found that the response to GSK126 treatment greatly varied between organoid lines. Response associated with the mutation status of ATRX and PAX2, and correlated with BIK expression. It also correlated well with response to Nutlin-3a which inhibits MDM2-p53 interaction thereby activating p53 signaling. Sensitivity to EZH2 ablation depended on the presence of wild type p53, as tumor organoids became resistant when p53 was mutated or knocked down. Our exploratory study provides insight into which genetic factors predict sensitivity to EZH2 inhibition. In addition, we show that the response to EZH2 inhibition requires wild type p53. We conclude that a subset of colorectal cancer patients may benefit from EZH2-targeting therapies.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Indoles/farmacología , Piridonas/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/análisis , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Proteína Potenciadora del Homólogo Zeste 2/análisis , Humanos , Proteínas de la Membrana/análisis , Ratones , Proteínas Mitocondriales , Mutación , Organoides , Factor de Transcripción PAX2/genética , Proteína p53 Supresora de Tumor/fisiología , Proteína Nuclear Ligada al Cromosoma X/genética
8.
Gastroenterology ; 151(4): 684-697.e12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27342214

RESUMEN

BACKGROUND & AIMS: The polycomb repressive complex 2 (PRC2) regulates differentiation by contributing to repression of gene expression and thereby stabilizing the fate of stem cells and their progeny. PRC2 helps to maintain adult stem cell populations, but little is known about its functions in intestinal stem cells. We studied phenotypes of mice with intestine-specific deletion of the PRC2 proteins embryonic ectoderm development (EED) (a subunit required for PRC2 function) and enhancer of zeste homolog 2 (EZH2) (a histone methyltransferase). METHODS: We performed studies of AhCre;EedLoxP/LoxP (EED knockout) mice and AhCre;Ezh2LoxP/LoxP (EZH2 knockout) mice, which have intestine-specific disruption in EED and EZH2, respectively. Small intestinal crypts were isolated and subsequently cultured to grow organoids. Intestines and organoids were analyzed by immunohistochemical, in situ hybridization, RNA sequence, and chromatin immunoprecipitation methods. RESULTS: Intestines of EED knockout mice had massive crypt degeneration and lower numbers of proliferating cells compared with wild-type control mice. Cdkn2a became derepressed and we detected increased levels of P21. We did not observe any differences between EZH2 knockout and control mice. Intestinal crypts from EED knockout mice had signs of aberrant differentiation of uncommitted crypt cells-these differentiated toward the secretory cell lineage. Furthermore, crypts from EED-knockout mice had impaired Wnt signaling and concomitant loss of intestinal stem cells, this phenotype was not reversed upon ectopic stimulation of Wnt and Notch signaling in organoids. Analysis of gene expression patterns from intestinal tissues of EED knockout mice showed dysregulation of several genes involved in Wnt signaling. Wnt signaling was regulated directly by PRC2. CONCLUSIONS: In intestinal tissues of mice, PRC2 maintains small intestinal stem cells by promoting proliferation and preventing differentiation in the intestinal stem cell compartment. PRC2 controls gene expression in multiple signaling pathways that regulate intestinal homeostasis. Sequencing data are available in the genomics data repository GEO under reference series GSE81578; RNA sequencing data are available under subseries GSE81576; and ChIP sequencing data are available under subseries GSE81577.


Asunto(s)
Células Madre Adultas/fisiología , Intestinos/citología , Complejo Represivo Polycomb 2/deficiencia , Animales , Secuencia de Bases , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Inmunoprecipitación de Cromatina , Proteína Potenciadora del Homólogo Zeste 2/deficiencia , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Complejo Represivo Polycomb 2/genética , Vía de Señalización Wnt
9.
Cancer Cell ; 29(1): 17-31, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26766588

RESUMEN

Polycomb repressive complexes (PRC) are frequently implicated in human cancer, acting either as oncogenes or tumor suppressors. Here, we show that PRC2 is a critical regulator of KRAS-driven non-small cell lung cancer progression. Modulation of PRC2 by either Ezh2 overexpression or Eed deletion enhances KRAS-driven adenomagenesis and inflammation, respectively. Eed-loss-driven inflammation leads to massive macrophage recruitment and marked decline in tissue function. Additional Trp53 inactivation activates a cell-autonomous epithelial-to-mesenchymal transition program leading to an invasive mucinous adenocarcinoma. A switch between methylated/acetylated chromatin underlies the tumor phenotypic evolution, prominently involving genes controlled by Hippo/Wnt signaling. Our observations in the mouse models were conserved in human cells. Importantly, PRC2 inactivation results in context-dependent phenotypic alterations, with implications for its therapeutic application.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Transición Epitelial-Mesenquimal/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Acetilación , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Histonas/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Ratones Transgénicos , Complejo Represivo Polycomb 2/genética , Proteínas Proto-Oncogénicas p21(ras)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...