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












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 5386, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918386

RESUMEN

Aberrantly accumulated metabolites elicit intra- and inter-cellular pro-oncogenic cascades, yet current measurement methods require sample perturbation/disruption and lack spatio-temporal resolution, limiting our ability to fully characterize their function and distribution. Here, we show that Raman spectroscopy (RS) can directly detect fumarate in living cells in vivo and animal tissues ex vivo, and that RS can distinguish between Fumarate hydratase (Fh1)-deficient and Fh1-proficient cells based on fumarate concentration. Moreover, RS reveals the spatial compartmentalization of fumarate within cellular organelles in Fh1-deficient cells: consistent with disruptive methods, we observe the highest fumarate concentration (37 ± 19 mM) in mitochondria, where the TCA cycle operates, followed by the cytoplasm (24 ± 13 mM) and then the nucleus (9 ± 6 mM). Finally, we apply RS to tissues from an inducible mouse model of FH loss in the kidney, demonstrating RS can classify FH status. These results suggest RS could be adopted as a valuable tool for small molecule metabolic imaging, enabling in situ non-destructive evaluation of fumarate compartmentalization.


Asunto(s)
Fumarato Hidratasa , Fumaratos , Espectrometría Raman , Espectrometría Raman/métodos , Animales , Fumaratos/metabolismo , Ratones , Fumarato Hidratasa/metabolismo , Fumarato Hidratasa/genética , Riñón/metabolismo , Mitocondrias/metabolismo , Humanos , Núcleo Celular/metabolismo , Citoplasma/metabolismo
2.
Nature ; 615(7952): 490-498, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890227

RESUMEN

Metabolic rewiring underlies the effector functions of macrophages1-3, but the mechanisms involved remain incompletely defined. Here, using unbiased metabolomics and stable isotope-assisted tracing, we show that an inflammatory aspartate-argininosuccinate shunt is induced following lipopolysaccharide stimulation. The shunt, supported by increased argininosuccinate synthase (ASS1) expression, also leads to increased cytosolic fumarate levels and fumarate-mediated protein succination. Pharmacological inhibition and genetic ablation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) further increases intracellular fumarate levels. Mitochondrial respiration is also suppressed and mitochondrial membrane potential increased. RNA sequencing and proteomics analyses demonstrate that there are strong inflammatory effects resulting from FH inhibition. Notably, acute FH inhibition suppresses interleukin-10 expression, which leads to increased tumour necrosis factor secretion, an effect recapitulated by fumarate esters. Moreover, FH inhibition, but not fumarate esters, increases interferon-ß production through mechanisms that are driven by mitochondrial RNA (mtRNA) release and activation of the RNA sensors TLR7, RIG-I and MDA5. This effect is recapitulated endogenously when FH is suppressed following prolonged lipopolysaccharide stimulation. Furthermore, cells from patients with systemic lupus erythematosus also exhibit FH suppression, which indicates a potential pathogenic role for this process in human disease. We therefore identify a protective role for FH in maintaining appropriate macrophage cytokine and interferon responses.


Asunto(s)
Fumarato Hidratasa , Interferón beta , Macrófagos , Mitocondrias , ARN Mitocondrial , Humanos , Argininosuccinato Sintasa/metabolismo , Ácido Argininosuccínico/metabolismo , Ácido Aspártico/metabolismo , Respiración de la Célula , Citosol/metabolismo , Fumarato Hidratasa/antagonistas & inhibidores , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Fumaratos/metabolismo , Interferón beta/biosíntesis , Interferón beta/inmunología , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Lupus Eritematoso Sistémico/enzimología , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Potencial de la Membrana Mitocondrial , Metabolómica , Mitocondrias/genética , Mitocondrias/metabolismo , ARN Mitocondrial/metabolismo
3.
Nature ; 615(7952): 499-506, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890229

RESUMEN

Mutations in fumarate hydratase (FH) cause hereditary leiomyomatosis and renal cell carcinoma1. Loss of FH in the kidney elicits several oncogenic signalling cascades through the accumulation of the oncometabolite fumarate2. However, although the long-term consequences of FH loss have been described, the acute response has not so far been investigated. Here we generated an inducible mouse model to study the chronology of FH loss in the kidney. We show that loss of FH leads to early alterations of mitochondrial morphology and the release of mitochondrial DNA (mtDNA) into the cytosol, where it triggers the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-TANK-binding kinase 1 (TBK1) pathway and stimulates an inflammatory response that is also partially dependent on retinoic-acid-inducible gene I (RIG-I). Mechanistically, we show that this phenotype is mediated by fumarate and occurs selectively through mitochondrial-derived vesicles in a manner that depends on sorting nexin 9 (SNX9). These results reveal that increased levels of intracellular fumarate induce a remodelling of the mitochondrial network and the generation of mitochondrial-derived vesicles, which allows the release of mtDNAin the cytosol and subsequent activation of the innate immune response.


Asunto(s)
ADN Mitocondrial , Fumaratos , Inmunidad Innata , Mitocondrias , Animales , Ratones , ADN Mitocondrial/metabolismo , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Fumaratos/metabolismo , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Riñón/enzimología , Riñón/metabolismo , Riñón/patología , Citosol/metabolismo
4.
Nat Commun ; 13(1): 7830, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36539415

RESUMEN

Metabolic reprogramming is critical for tumor initiation and progression. However, the exact impact of specific metabolic changes on cancer progression is poorly understood. Here, we integrate multimodal analyses of primary and metastatic clonally-related clear cell renal cancer cells (ccRCC) grown in physiological media to identify key stage-specific metabolic vulnerabilities. We show that a VHL loss-dependent reprogramming of branched-chain amino acid catabolism sustains the de novo biosynthesis of aspartate and arginine enabling tumor cells with the flexibility of partitioning the nitrogen of the amino acids depending on their needs. Importantly, we identify the epigenetic reactivation of argininosuccinate synthase (ASS1), a urea cycle enzyme suppressed in primary ccRCC, as a crucial event for metastatic renal cancer cells to acquire the capability to generate arginine, invade in vitro and metastasize in vivo. Overall, our study uncovers a mechanism of metabolic flexibility occurring during ccRCC progression, paving the way for the development of novel stage-specific therapies.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/genética , Aminoácidos de Cadena Ramificada , Nitrógeno , Neoplasias Renales/genética , Arginina/metabolismo , Línea Celular Tumoral
5.
Oncogene ; 38(7): 1136-1150, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30237440

RESUMEN

Elucidation of mechanisms underlying the increased androgen receptor (AR) activity and subsequent development of aggressive prostate cancer (PrCa) is pivotal in developing new therapies. Using a systems biology approach, we interrogated the AR-regulated proteome and identified PDZ binding kinase (PBK) as a novel AR-regulated protein that regulates full-length AR and AR variants (ARVs) activity in PrCa. PBK overexpression in aggressive PrCa is associated with early biochemical relapse and poor clinical outcome. In addition to its carboxy terminus ligand-binding domain, PBK directly interacts with the amino terminus transactivation domain of the AR to stabilise it thereby leading to increased AR protein expression observed in PrCa. Transcriptome sequencing revealed that PBK is a mediator of global AR signalling with key roles in regulating tumour invasion and metastasis. PBK inhibition decreased growth of PrCa cell lines and clinical specimen cultured ex vivo. We uncovered a novel interplay between AR and PBK that results in increased AR and ARVs expression that executes AR-mediated growth and progression of PrCa, with implications for the development of PBK inhibitors for the treatment of aggressive PrCa.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Inhibidores de Proteínas Quinasas/farmacología , Receptores Androgénicos/genética
6.
Nat Commun ; 8(1): 374, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851861

RESUMEN

Emerging data demonstrate homologous recombination (HR) defects in castration-resistant prostate cancers, rendering these tumours sensitive to PARP inhibition. Here we demonstrate a direct requirement for the androgen receptor (AR) to maintain HR gene expression and HR activity in prostate cancer. We show that PARP-mediated repair pathways are upregulated in prostate cancer following androgen-deprivation therapy (ADT). Furthermore, upregulation of PARP activity is essential for the survival of prostate cancer cells and we demonstrate a synthetic lethality between ADT and PARP inhibition in vivo. Our data suggest that ADT can functionally impair HR prior to the development of castration resistance and that, this potentially could be exploited therapeutically using PARP inhibitors in combination with androgen-deprivation therapy upfront in advanced or high-risk prostate cancer.Tumours with homologous recombination (HR) defects become sensitive to PARPi. Here, the authors show that androgen receptor (AR) regulates HR and AR inhibition activates the PARP pathway in vivo, thus inhibition of both AR and PARP is required for effective treatment of high risk prostate cancer.


Asunto(s)
Colágeno Tipo XI/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/metabolismo , Mutaciones Letales Sintéticas , Colágeno Tipo XI/genética , Recombinación Homóloga , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Transducción de Señal
7.
Int J Cancer ; 140(8): 1881-1887, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28073170

RESUMEN

We have previously reported that the negative signaling regulator Similar Expression to FGF (hSef) is downregulated in prostate cancer and its loss is associated with clinical metastasis. Here, we explored the mechanistic basis of this finding. We first confirmed our clinical observation by testing hSef manipulation in an in vivo metastasis model. hSef stable expressing cells (PC3M-hSef) or empty vector controls (PC3M-EV) were injected subcutaneously into the lateral thoracic walls of NOD-SCID gamma mice and lungs were harvested at autopsy. In this model, 6/7 PC3M-EV xenografts had definitive lung micro-metastasis whilst only 1/6 PC3M-hSef xenografts exhibited metastasis recapitulating the clinical scenario (p = 0.03). Gene expression studies revealed key perturbations in genes involved in cell motility and epithelial to mesenchymal transition (EMT) along with alterations in cognate signaling pathways. These results were validated in an EMT specific PCR array whereby hSef over-expression and silencing reciprocally altered E-Cadherin expression (p = <0.001) amongst other EMT markers. Immunohistochemistry of excised tumors from the xenografts also confirmed the effect of hSef in suppressing E-Cadherin expression at the protein level. Phosphokinase arrays further demonstrated a role for hSef in attenuating signaling of not only ERK-MAPK but also the JNK and p38 pathways as well. Taken together, these data suggest evidence that loss of hSef may be a critical event facilitating tumor dissemination of prostate cancer through alteration of EMT. Detection of downregulated hSef, along with other negative regulators, may therefore be a useful biomarker heralding a transition to a metastatic phenotype and warrants further exploration in this context.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Transición Epitelial-Mesenquimal/genética , Neoplasias de la Próstata/genética , Receptores de Interleucina/genética , Animales , Antígenos CD , Biomarcadores de Tumor/genética , Cadherinas/biosíntesis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis
8.
Biochim Biophys Acta Bioenerg ; 1858(8): 723-731, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27956047

RESUMEN

Our understanding of cancer has recently seen a major paradigm shift resulting in it being viewed as a metabolic disorder, and altered cellular metabolism being recognised as a hallmark of cancer. This concept was spurred by the findings that the oncogenic mutations driving tumorigenesis induce a reprogramming of cancer cell metabolism that is required for unrestrained growth and proliferation. The recent discovery that mutations in key mitochondrial enzymes play a causal role in tumorigenesis suggested that dysregulation of metabolism could also be a driver of tumorigenesis. These mutations induce profound adaptive metabolic alterations that are a prerequisite for the survival of the mutated cells. Because these metabolic events are specific to cancer cells, they offer an opportunity to develop new therapies that specifically target tumour cells without affecting healthy tissue. Here, we will describe recent developments in metabolism-based cancer therapy, in particular focusing on the concept of metabolic synthetic lethality. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Metabolismo Energético/efectos de los fármacos , Proteínas de Neoplasias/genética , Neoplasias/tratamiento farmacológico , Mutaciones Letales Sintéticas , Antimetabolitos Antineoplásicos/uso terapéutico , Simulación por Computador , Metabolismo Energético/genética , Predicción , Dosificación de Gen , Silenciador del Gen , Genes Relacionados con las Neoplasias , Humanos , Metaboloma , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Modelos Biológicos , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Oncogenes , Fosforilación Oxidativa , Interferencia de ARN
10.
Nature ; 537(7621): 544-547, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27580029

RESUMEN

Mutations of the tricarboxylic acid cycle enzyme fumarate hydratase cause hereditary leiomyomatosis and renal cell cancer. Fumarate hydratase-deficient renal cancers are highly aggressive and metastasize even when small, leading to a very poor clinical outcome. Fumarate, a small molecule metabolite that accumulates in fumarate hydratase-deficient cells, plays a key role in cell transformation, making it a bona fide oncometabolite. Fumarate has been shown to inhibit α-ketoglutarate-dependent dioxygenases that are involved in DNA and histone demethylation. However, the link between fumarate accumulation, epigenetic changes, and tumorigenesis is unclear. Here we show that loss of fumarate hydratase and the subsequent accumulation of fumarate in mouse and human cells elicits an epithelial-to-mesenchymal-transition (EMT), a phenotypic switch associated with cancer initiation, invasion, and metastasis. We demonstrate that fumarate inhibits Tet-mediated demethylation of a regulatory region of the antimetastatic miRNA cluster mir-200ba429, leading to the expression of EMT-related transcription factors and enhanced migratory properties. These epigenetic and phenotypic changes are recapitulated by the incubation of fumarate hydratase-proficient cells with cell-permeable fumarate. Loss of fumarate hydratase is associated with suppression of miR-200 and the EMT signature in renal cancer and is associated with poor clinical outcome. These results imply that loss of fumarate hydratase and fumarate accumulation contribute to the aggressive features of fumarate hydratase-deficient tumours.


Asunto(s)
Epigénesis Genética , Transición Epitelial-Mesenquimal , Fumaratos/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Células HEK293 , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Mesodermo/metabolismo , Ratones , MicroARNs/genética , Factores de Transcripción/metabolismo , Transcriptoma
11.
J Natl Cancer Inst ; 108(5)2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26657335

RESUMEN

BACKGROUND: The androgen receptor (AR) is a major drug target in prostate cancer (PCa). We profiled the AR-regulated kinome to identify clinically relevant and druggable effectors of AR signaling. METHODS: Using genome-wide approaches, we interrogated all AR regulated kinases. Among these, choline kinase alpha (CHKA) expression was evaluated in benign (n = 195), prostatic intraepithelial neoplasia (PIN) (n = 153) and prostate cancer (PCa) lesions (n = 359). We interrogated how CHKA regulates AR signaling using biochemical assays and investigated androgen regulation of CHKA expression in men with PCa, both untreated (n = 20) and treated with an androgen biosynthesis inhibitor degarelix (n = 27). We studied the effect of CHKA inhibition on the PCa transcriptome using RNA sequencing and tested the effect of CHKA inhibition on cell growth, clonogenic survival and invasion. Tumor xenografts (n = 6 per group) were generated in mice using genetically engineered prostate cancer cells with inducible CHKA knockdown. Data were analyzed with χ(2) tests, Cox regression analysis, and Kaplan-Meier methods. All statistical tests were two-sided. RESULTS: CHKA expression was shown to be androgen regulated in cell lines, xenografts, and human tissue (log fold change from 6.75 to 6.59, P = .002) and was positively associated with tumor stage. CHKA binds directly to the ligand-binding domain (LBD) of AR, enhancing its stability. As such, CHKA is the first kinase identified as an AR chaperone. Inhibition of CHKA repressed the AR transcriptional program including pathways enriched for regulation of protein folding, decreased AR protein levels, and inhibited the growth of PCa cell lines, human PCa explants, and tumor xenografts. CONCLUSIONS: CHKA can act as an AR chaperone, providing, to our knowledge, the first evidence for kinases as molecular chaperones, making CHKA both a marker of tumor progression and a potential therapeutic target for PCa.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Colina Quinasa/metabolismo , Chaperonas Moleculares , Terapia Molecular Dirigida/métodos , Prostatectomía , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Receptores Androgénicos/metabolismo , Transducción de Señal , Anciano , Animales , Colina Quinasa/antagonistas & inhibidores , Colina Quinasa/genética , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Modelos de Riesgos Proporcionales , Prostatectomía/métodos , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Análisis de Secuencia de ADN , Ensayos Antitumor por Modelo de Xenoinjerto
12.
EMBO J ; 33(12): 1365-82, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24837709

RESUMEN

Tumour cells sustain their high proliferation rate through metabolic reprogramming, whereby cellular metabolism shifts from oxidative phosphorylation to aerobic glycolysis, even under normal oxygen levels. Hypoxia-inducible factor 1A (HIF1A) is a major regulator of this process, but its activation under normoxic conditions, termed pseudohypoxia, is not well documented. Here, using an integrative approach combining the first genome-wide mapping of chromatin binding for an endocytic adaptor, ARRB1, both in vitro and in vivo with gene expression profiling, we demonstrate that nuclear ARRB1 contributes to this metabolic shift in prostate cancer cells via regulation of HIF1A transcriptional activity under normoxic conditions through regulation of succinate dehydrogenase A (SDHA) and fumarate hydratase (FH) expression. ARRB1-induced pseudohypoxia may facilitate adaptation of cancer cells to growth in the harsh conditions that are frequently encountered within solid tumours. Our study is the first example of an endocytic adaptor protein regulating metabolic pathways. It implicates ARRB1 as a potential tumour promoter in prostate cancer and highlights the importance of metabolic alterations in prostate cancer.


Asunto(s)
Arrestinas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Redes y Vías Metabólicas/fisiología , Modelos Biológicos , Neoplasias de la Próstata/fisiopatología , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Fumarato Hidratasa/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Espectroscopía de Resonancia Magnética , Masculino , Metabolómica , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , Succinato Deshidrogenasa/metabolismo , Análisis de Matrices Tisulares , beta-Arrestina 1 , beta-Arrestinas
14.
Cancer Cell ; 23(1): 35-47, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23260764

RESUMEN

The androgen receptor (AR) regulates prostate cell growth in man, and prostate cancer is the commonest cancer in men in the UK. We present a comprehensive analysis of AR binding sites in human prostate cancer tissues, including castrate-resistant prostate cancer (CRPC). We identified thousands of AR binding sites in CRPC tissue, most of which were not identified in PC cell lines. Many adjacent genes showed AR regulation in xenografts but not in cultured LNCaPs, demonstrating an in-vivo-restricted set of AR-regulated genes. Functional studies support a model of altered signaling in vivo that directs AR binding. We identified a 16 gene signature that outperformed a larger in-vitro-derived signature in clinical data sets, showing the importance of persistent AR signaling in CRPC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Receptores Androgénicos/fisiología , Animales , Sitios de Unión , Línea Celular Tumoral , Histonas/metabolismo , Humanos , Masculino , Ratones , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo
15.
PLoS One ; 6(6): e20767, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698231

RESUMEN

Here we investigate the effects of expressing an activated mutant of Notch (ICD-E) in an inducible transgenic mouse model. Hepatic expression of ICD-E in adult animals has no detectable phenotype, but simultaneous induction of ICD-E in both the liver and small intestine results in hepatic steatosis, lipogranuloma formation and mild insulin resistance within 96 hours. This supports work that suggests that fatty liver disease may result from disruption of the gut-liver axis. In the intestine, ICD-E expression is known to produce a transient change in the proportion of goblet cells followed by shedding of the recombinant epithelium. We report additional intestinal transcriptional changes following ICD-E expression, finding significant transcriptional down-regulation of rpL29 (ribosomal protein L29), which is implicated in the regulation of intestinal flora. These results provide further evidence of a gut-liver axis in the development of fatty liver disease and insulin resistance and validate a new model for future studies of hepatic steatosis.


Asunto(s)
Hígado Graso/metabolismo , Resistencia a la Insulina , Mucosa Intestinal/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ribosómicas/genética , Transcripción Genética
16.
Traffic ; 10(9): 1209-20, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19453971

RESUMEN

A subset of proteins predominantly associated with early endosomes or implicated in clathrin-mediated endocytosis can shuttle between the cytoplasm and the nucleus. Although the endocytic functions of these proteins have been extensively studied, much less effort has been expended in exploring their nuclear roles. Membrane trafficking proteins can affect signalling and proliferation and this can be achieved either at a nuclear or endocytic level. Furthermore, some proteins, such as Huntingtin interacting protein 1, are known as cancer biomarkers. This review will highlight the limits of our understanding of their nuclear functions and the relevance of this to signalling and oncogenesis.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Endosomas/metabolismo , Neoplasias/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Endocitosis , Humanos , Neoplasias/genética
17.
J Cell Biochem ; 107(1): 19-29, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19204937

RESUMEN

Chromatin immunoprecipitation (ChIP), when paired with sequencing or arrays, has become a method of choice for the unbiased identification of genomic-binding sites for transcription factors and epigenetic marks in various model systems. The data generated is often then interpreted by groups seeking to link these binding sites to the expression of adjacent or distal genes, and more broadly to the evolution of species, cell fate/differentiation or even cancer development. Against this backdrop is an ongoing debate over the relative importance DNA sequence versus chromatin structure and modification in the regulation of gene expression (Anon. 2008a Nature 454: 795; Anon. 2008b Nature 454: 711-715; Henikoff et al. 2008 Science 322: 853; Madhani et al. 2008 Science 322: 43-44). Rationally there is a synergy between the two and the goal of a biologist is to characterise both comprehensively enough to explain a cellular phenotype or a developmental process. If this is truly our goal then the critical factor in good science is an awareness of the constraints and potential of the biological models used. The reality however is often that this discussion is polarised by funding imperatives and the need to align to a transcription factor or epigenetic camp. This article will discuss the extrapolations involved in using ChIP data to draw conclusions about these themes and the discoveries that have resulted.


Asunto(s)
Inmunoprecipitación de Cromatina , Epigénesis Genética , Regulación de la Expresión Génica , Factores de Transcripción , Transcripción Genética , Animales , Humanos
18.
Dev Dyn ; 235(4): 998-1013, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16534797

RESUMEN

The Notch gene of Drosophila encodes a single transmembrane receptor that plays a central role in the process of lateral inhibition. This process results in the selection of individual mesodermal and neural precursors during the development of the muscular and nervous systems. The activation of Notch during lateral inhibition is mediated by the transmembrane ligand Delta (Dl) and effected by the transcription factor Suppressor of Hairless (Su(H)). The same functional cassette plays a role in other processes, in particular, the development and patterning of the wing. Genetic analysis has suggested that, in addition to the Su(H)-dependent pathway, Notch can signal in an Su(H)-independent manner. This process seems to be tightly associated with signalling by Wingless, a member of the Wnt family of signalling molecules. Here, we have analyzed further the possibility that the Notch protein encodes two different functions. To do so, we have studied the activities and genetic properties of different Notch receptors bearing deletions of specific regions of the intracellular and the extracellular domains in different developmental processes, and have sought to correlate the activity of these mutant proteins with those of existing mutants in Notch. Our results support the existence of at least two different activities of Notch each of which can be associated with specific structural domains.


Asunto(s)
Tipificación del Cuerpo , Drosophila/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Animales , Drosophila/embriología , Embrión no Mamífero , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ingeniería Genética , Estructura Terciaria de Proteína , Receptores Notch/química , Transgenes , Alas de Animales/embriología
19.
Genes Dev ; 19(14): 1686-91, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024658

RESUMEN

The intestinal epithelium comprises differentiated cells of four lineages maintained by precursor cells. As the Notch pathway controls the fate of proliferating cells in many systems, we investigated the effect of conditional expression of an activated Notch mutant in intestinal epithelium. An increase in the number of goblet cells occurs within 8 h of induction, due to an effect of Notch on post-mitotic cells, not on precursors. This observation broadens the role of Notch into controlling postmitotic differentiation and indicates that the composition of the epithelium is not solely determined by progenitor cells.


Asunto(s)
Proteínas de Drosophila/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Diferenciación Celular , Línea Celular , Cricetinae , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Caliciformes/citología , Células Caliciformes/metabolismo , Humanos , Inmunohistoquímica , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Ratones Transgénicos , Mitosis , Ratas , Receptores Notch , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección
20.
Curr Opin Genet Dev ; 12(5): 524-33, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12200157

RESUMEN

During development, signalling by members of the Notch family of cell surface receptors plays a widespread role in the assignation of cell fates within the process of lateral inhibition. This function of Notch is mediated by a well-established mechanism that relies on a ligand-induced release of the intracellular domain of Notch (NICD) and the interaction of this fragment with members of the CSL (CBF1, Suppressor of Hairless, Lag-1) family of transcription factors within the nucleus. However, there is increasing evidence that Notch can signal in CSL-independent modes.


Asunto(s)
Ciclo Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Alas de Animales/embriología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila , Drosophila melanogaster/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Receptores Notch , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...