Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 10(8)2021 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-34440795

RESUMEN

During liver organogenesis, cellular transcriptional profiles are constantly reshaped by the action of hepatic transcriptional regulators, including FoxA1-3, GATA4/6, HNF1α/ß, HNF4α, HNF6, OC-2, C/EBPα/ß, Hex, and Prox1. These factors are crucial for the activation of hepatic genes that, in the context of compact chromatin, cannot access their targets. The initial opening of highly condensed chromatin is executed by a special class of transcription factors known as pioneer factors. They bind and destabilize highly condensed chromatin and facilitate access to other "non-pioneer" factors. The association of target genes with pioneer and non-pioneer transcription factors takes place long before gene activation. In this way, the underlying gene regulatory regions are marked for future activation. The process is called "bookmarking", which confers transcriptional competence on target genes. Developmental bookmarking is accompanied by a dynamic maturation process, which prepares the genomic loci for stable and efficient transcription. Stable hepatic expression profiles are maintained during development and adulthood by the constant availability of the main regulators. This is achieved by a self-sustaining regulatory network that is established by complex cross-regulatory interactions between the major regulators. This network gradually grows during liver development and provides an epigenetic memory mechanism for safeguarding the optimal expression of the regulators.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hígado/metabolismo , Organogénesis/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/embriología , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo
2.
iScience ; 24(5): 102473, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34113819

RESUMEN

The oncogenic function of suppressor of variegation, enhancer of zeste and MYeloid-Nervy-DEAF1-domain family methyltransferase Smyd3 has been implicated in various malignancies, including hepatocellular carcinoma (HCC). Here, we show that targeting Smyd3 by next-generation antisense oligonucleotides (Smyd3-ASO) is an efficient approach to modulate its mRNA levels in vivo and to halt the growth of already initiated liver tumors. Smyd3-ASO treatment dramatically decreased tumor burden in a mouse model of chemically induced HCC and negatively affected the growth rates, migration, oncosphere formation, and xenograft growth capacity of a panel of human hepatic cancer cell lines. Smyd3-ASOs prevented the activation of oncofetal genes and the development of cancer-specific gene expression program. The results point to a mechanism by which Smyd3-ASO treatment blocks cellular de-differentiation, a hallmark feature of HCC development, and, as a result, it inhibits the expansion of hepatic cancer stem cells, a population that has been presumed to resist chemotherapy.

3.
EMBO J ; 40(1): e102236, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33034061

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) from somatic cells provides an excellent model to study mechanisms of transcription factor-induced global alterations of the epigenome and genome function. Here, we have investigated the early transcriptional events of cellular reprogramming triggered by the co-expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) in mouse embryonic fibroblasts (MEFs) and mouse hepatocytes (mHeps). In this analysis, we identified a gene regulatory network composed of nine transcriptional regulators (9TR; Cbfa2t3, Gli2, Irf6, Nanog, Ovol1, Rcan1, Taf1c, Tead4, and Tfap4), which are directly targeted by OSKM, in vivo. Functional studies using single and double shRNA knockdowns of any of these factors caused disruption of the network and dramatic reductions in reprogramming efficiency, indicating that this network is essential for the induction and establishment of pluripotency. We demonstrate that the stochastic co-expression of 9TR network components occurs in a remarkably small number of cells, approximating the percentage of terminally reprogrammed cells as a result of dynamic molecular events. Thus, the early DNA-binding patterns of OSKM and the subsequent probabilistic co-expression of essential 9TR components in subpopulations of cells undergoing reprogramming steer the reconstruction of a gene regulatory network marking the transition to pluripotency.


Asunto(s)
Reprogramación Celular/genética , Fibroblastos/fisiología , Redes Reguladoras de Genes/genética , Hepatocitos/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Animales , Células Madre Embrionarias/fisiología , Femenino , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Embarazo , Factores de Transcripción/genética , Transcripción Genética/genética
4.
Cell Rep ; 30(5): 1319-1328.e6, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32023452

RESUMEN

Transcription factor binding to enhancer and promoter regions critical for homeostatic adult gene activation is established during development. To understand how cell-specific gene expression patterns are generated, we study the developmental timing of association of two prominent hepatic transcription factors with gene regulatory regions. Most individual binding events display extraordinarily high temporal variations during liver development. Early and persistent binding is necessary, but not sufficient, for gene activation. Stable gene expression patterns are the result of combinatorial activity of multiple transcription factors, which mark regulatory regions long before activation and promote progressive broadening of active chromatin domains. Both temporally stable and dynamic, short-lived binding events contribute to the developmental maturation of active promoter configurations. The results reveal a developmental bookmarking function of master regulators and illuminate remarkable parallels between the principles employed for gene activation during development, during evolution, and upon mitotic exit.


Asunto(s)
Hígado/embriología , Hígado/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos
5.
Cell Rep ; 20(4): 909-922, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28746875

RESUMEN

H4K20 monomethylation maintains genome integrity by regulating proper mitotic condensation, DNA damage response, and replication licensing. Here, we show that, in non-dividing hepatic cells, H4K20Me1 is specifically enriched in active gene bodies and dynamically regulated by the antagonistic action of Kmt5a methylase and Kdm7b demethylase. In liver-specific Kmt5a-deficient mice, reduced levels of H4K20Me1 correlated with reduced RNA Pol II release from promoter-proximal regions. Genes regulating glucose and fatty acid metabolism were most sensitive to impairment of RNA Pol II release. Downregulation of glycolytic genes resulted in an energy starvation condition partially compensated by AMP-activated protein kinase (AMPK) activation and increased mitochondrial activity. This metabolic reprogramming generated a highly sensitized state that, upon different metabolic stress conditions, quickly aggravated into a senescent phenotype due to ROS overproduction-mediated oxidative DNA damage. The results illustrate how defects in the general process of RNA Pol II transition into a productive elongation phase can trigger specific metabolic changes and genome instability.


Asunto(s)
Histonas/metabolismo , Hígado/metabolismo , Proteína Metiltransferasas/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética/genética , Animales , Regulación de la Expresión Génica , Histonas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Proteína Metiltransferasas/genética , ARN Polimerasa II/genética
6.
ACS Chem Biol ; 12(3): 654-663, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28059499

RESUMEN

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII/NR2F2) is an orphan member of the nuclear receptor family of transcription factors whose activities are modulated upon binding of small molecules into an hydrophobic ligand-binding pocket (LBP). Although the LBP of COUP-TFII is filled with aromatic amino-acid side chains, alternative modes of ligand binding could potentially lead to regulation of the orphan receptor. Here, we screened a synthetic and natural compound library in a yeast one-hybrid assay and identified 4-methoxynaphthol as an inhibitor of COUP-TFII. This synthetic inhibitor was able to counteract processes either positively or negatively regulated by COUP-TFII in different mammalian cell systems. Hence, we demonstrate that the true orphan receptor COUP-TFII can be targeted by small chemicals which could be used to study the physiological functions of COUP-TFII or to counteract detrimental COUP-TFII activities in various pathological conditions.


Asunto(s)
Factor de Transcripción COUP II/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Sitios de Unión , Factor de Transcripción COUP II/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Hep G2 , Humanos , Ratones
7.
Semin Cancer Biol ; 42: 70-80, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27554136

RESUMEN

SMYD3 is a member of the SET and MYND-domain family of methyl-transferases, the increased expression of which correlates with poor prognosis in various types of cancer. In liver and colon tumors, SMYD3 is localized in the nucleus, where it interacts with RNA Pol II and H3K4me3 and functions as a selective transcriptional amplifier of oncogenes and genes that control cell proliferation and metastatic spread. Smyd3 expression has a high discriminative power for the characterization of liver tumors and positively correlates with poor prognosis. In lung and pancreatic cancer, SMYD3 acts in the cytoplasm, potentiating oncogenic Ras/ERK signaling through the methylation of the MAP3K2 kinase and the subsequent release from its inhibitor. A clinico-pathological analysis of lung cancer patients uncovers prognostic significance of SMYD3 only for first progression survival. However, stratification of patients according to their smoking history significantly expands the prognostic value of SMYD3 to overall survival and other features, suggesting that smoking-related effects saturate the clinical analysis and mask the function of SMYD3 as an oncogenic potentiator.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Neoplasias/genética , Pronóstico , Carcinogénesis/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/biosíntesis , Humanos , Neoplasias/patología , ARN Polimerasa II/genética
8.
Mol Cell Oncol ; 3(1): e1021946, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27308536

RESUMEN

Cancer stem cells (CSCs) are defined as cells within tumors that can self-renew and differentiate into heterogeneous lineages of cancerous cells. The origin of CSCs is not well understood. Recent evidence suggests that CSCs in hepatocellular carcinoma could be generated via oncogenic transformation and partial differentiation of adult hepatic ductal progenitor cells.

10.
Cell Rep ; 15(12): 2733-44, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27292644

RESUMEN

TGF-ß signaling regulates a variety of cellular processes, including proliferation, apoptosis, differentiation, immune responses, and fibrogenesis. Here, we describe a lysine methylation-mediated mechanism that controls the pro-fibrogenic activity of TGF-ß. We find that the methyltransferase Set9 potentiates TGF-ß signaling by targeting Smad7, an inhibitory downstream effector. Smad7 methylation promotes interaction with the E3 ligase Arkadia and, thus, ubiquitination-dependent degradation. Depletion or pharmacological inhibition of Set9 results in elevated Smad7 protein levels and inhibits TGF-ß-dependent expression of genes encoding extracellular matrix components. The inhibitory effect of Set9 on TGF-ß-mediated extracellular matrix production is further demonstrated in mouse models of pulmonary fibrosis. Lung fibrosis induced by bleomycin or Ad-TGF-ß treatment was highly compromised in Set9-deficient mice. These results uncover a complex regulatory interplay among multiple Smad7 modifications and highlight the possibility that protein methyltransferases may represent promising therapeutic targets for treating lung fibrosis.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína Metiltransferasas/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Acetilación , Animales , Bleomicina , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Lisina/metabolismo , Masculino , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas Nucleares/metabolismo , Estabilidad Proteica , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Transducción de Señal/genética , Proteína smad7/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética
11.
EMBO Rep ; 17(5): 769-79, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26993089

RESUMEN

Pioneering studies within the last few years have allowed the in vitro expansion of tissue-specific adult stem cells from a variety of endoderm-derived organs, including the stomach, small intestine, and colon. Expansion of these cells requires activation of the receptor Lgr5 by its ligand R-spondin 1 and is likely facilitated by the fact that in healthy adults the stem cells in these organs are highly proliferative. In many other adult organs, such as the liver, proliferating cells are normally not abundant in adulthood. However, upon injury, the liver has a strong regenerative potential that is accompanied by the emergence of Lgr5-positive stem cells; these cells can be isolated and expanded in vitro as organoids. In an effort to isolate stem cells from non-regenerating mouse livers, we discovered that healthy gallbladders are a rich source of stem/progenitor cells that can be propagated in culture as organoids for more than a year. Growth of these organoids was stimulated by R-spondin 1 and noggin, whereas in the absence of these growth factors, the organoids differentiated partially toward the hepatocyte fate. When transplanted under the liver capsule, gallbladder-derived organoids maintained their architecture for 2 weeks. Furthermore, single cells prepared from dissociated organoids and injected into the mesenteric vein populated the liver parenchyma of carbon tetrachloride-treated mice. Human gallbladders were also a source of organoid-forming stem cells. Thus, under specific growth conditions, stem cells can be isolated from healthy gallbladders, expanded almost indefinitely in vitro, and induced to differentiate toward the hepatocyte lineage.


Asunto(s)
Proteínas Portadoras/metabolismo , Vesícula Biliar/citología , Células Madre/metabolismo , Trombospondinas/metabolismo , Animales , Biomarcadores , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Diferenciación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Hígado/citología , Ratones , Ratones Transgénicos , Organoides , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Células Madre/efectos de los fármacos , Trombospondinas/genética , Trombospondinas/farmacología , Transcriptoma
12.
Cancer Cell ; 29(3): 354-366, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26908355

RESUMEN

Smyd3 is a protein methyltransferase implicated in cancer development. Here we show that Smyd3 expression in mice is required for chemically induced liver and colon cancer formation. In these organs Smyd3 functions in the nucleus, stimulating the transcription of several key regulators involved in cell proliferation, epithelial-mesenchymal transition, the JAK/Stat3 oncogenic pathway, as well as the Myc and Ctnnb1 oncogenes. Smyd3 interacts with H3K4Me3-modified histone tails, which facilitates its recruitment to the core promoter regions of most active genes. Smyd3 binding density on target genes positively correlates with increased RNA polymerase-II density and transcriptional outputs. Despite its widespread distribution, the transcription-potentiating function of Smyd3 is restricted to a particular set of genes, whose expression is induced specifically during carcinogenesis.


Asunto(s)
Neoplasias del Colon/genética , N-Metiltransferasa de Histona-Lisina/genética , Neoplasias Hepáticas/genética , Transcripción Genética/genética , Animales , Carcinogénesis/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Quinasas Janus/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-myc/genética , ARN Polimerasa II/genética , Factor de Transcripción STAT3/genética , Activación Transcripcional/genética , beta Catenina/genética
13.
EMBO J ; 34(4): 430-47, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25515659

RESUMEN

PR-SET7-mediated histone 4 lysine 20 methylation has been implicated in mitotic condensation, DNA damage response and replication licensing. Here, we show that PR-SET7 function in the liver is pivotal for maintaining genome integrity. Hepatocyte-specific deletion of PR-SET7 in mouse embryos resulted in G2 phase arrest followed by massive cell death and defect in liver organogenesis. Inactivation at postnatal stages caused cell duplication-dependent hepatocyte necrosis, accompanied by inflammation, fibrosis and compensatory growth induction of neighboring hepatocytes and resident ductal progenitor cells. Prolonged necrotic regenerative cycles coupled with oncogenic STAT3 activation led to the spontaneous development of hepatic tumors composed of cells with cancer stem cell characteristics. These include a capacity to self-renew in culture or in xenografts and the ability to differentiate to phenotypically distinct hepatic cells. Hepatocellular carcinoma in PR-SET7-deficient mice displays a cancer stem cell gene signature specified by the co-expression of ductal progenitor markers and oncofetal genes.


Asunto(s)
Carcinoma Hepatocelular/enzimología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Células Madre Neoplásicas/enzimología , Animales , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/metabolismo , Masculino , Metilación , Ratones , Células Tumorales Cultivadas
14.
Biochim Biophys Acta ; 1851(1): 2-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24814048

RESUMEN

Tight regulation of lipid levels is critical for cellular and organismal homeostasis, not only in terms of energy utilization and storage, but also to prevent potential toxicity. The liver utilizes a set of hepatic transcription factors to regulate the expression of genes implicated in all aspects of lipid metabolism including catabolism, transport, and synthesis. In this article, we will review the main transcriptional mechanisms regulating the expression of genes involved in hepatic lipid metabolism. The principal regulatory pathways are composed of simple modules of transcription factor crosstalks, which correspond to building blocks of more complex regulatory networks. These transcriptional networks contribute to the regulation of proper lipid homeostasis in parallel to posttranslational mechanisms and end product-mediated modulation of lipid metabolizing enzymes. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.


Asunto(s)
Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Animales
15.
Genome Res ; 24(11): 1797-807, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25122613

RESUMEN

The genetic code is an abstraction of how mRNA codons and tRNA anticodons molecularly interact during protein synthesis; the stability and regulation of this interaction remains largely unexplored. Here, we characterized the expression of mRNA and tRNA genes quantitatively at multiple time points in two developing mouse tissues. We discovered that mRNA codon pools are highly stable over development and simply reflect the genomic background; in contrast, precise regulation of tRNA gene families is required to create the corresponding tRNA transcriptomes. The dynamic regulation of tRNA genes during development is controlled in order to generate an anticodon pool that closely corresponds to messenger RNAs. Thus, across development, the pools of mRNA codons and tRNA anticodons are invariant and highly correlated, revealing a stable molecular interaction interlocking transcription and translation.


Asunto(s)
Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hígado/metabolismo , ARN Mensajero/genética , ARN de Transferencia/genética , Transcriptoma , Animales , Anticodón/genética , Secuencia de Bases , Encéfalo/embriología , Inmunoprecipitación de Cromatina/métodos , Codón/genética , Simulación por Computador , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Hígado/embriología , Masculino , Ratones Endogámicos C57BL , Modelos Genéticos , Sistemas de Lectura Abierta/genética , Análisis de Componente Principal , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Factores de Tiempo
16.
Nat Commun ; 5: 4093, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912735

RESUMEN

Exposure to environmental cues such as cold or nutritional imbalance requires white adipose tissue (WAT) to adapt its metabolism to ensure survival. Metabolic plasticity is prominently exemplified by the enhancement of mitochondrial biogenesis in WAT in response to cold exposure or ß3-adrenergic stimulation. Here we show that these stimuli increase the levels of lysine-specific demethylase 1 (LSD1) in WAT of mice and that elevated LSD1 levels induce mitochondrial activity. Genome-wide binding and transcriptome analyses demonstrate that LSD1 directly stimulates the expression of genes involved in oxidative phosphorylation (OXPHOS) in cooperation with nuclear respiratory factor 1 (Nrf1). In transgenic (Tg) mice, increased levels of LSD1 promote in a cell-autonomous manner the formation of islets of metabolically active brown-like adipocytes in WAT. Notably, Tg mice show limited weight gain when fed a high-fat diet. Taken together, our data establish LSD1 as a key regulator of OXPHOS and metabolic adaptation in WAT.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Blanco/metabolismo , Frío , Histona Demetilasas/genética , Mitocondrias/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Fosforilación Oxidativa , Animales , Dieta Alta en Grasa , Metabolismo Energético , Humanos , Ratones , Ratones Transgénicos , Receptores Adrenérgicos beta 3/metabolismo , Transducción de Señal
17.
Cell ; 154(3): 530-40, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911320

RESUMEN

To mechanistically characterize the microevolutionary processes active in altering transcription factor (TF) binding among closely related mammals, we compared the genome-wide binding of three tissue-specific TFs that control liver gene expression in six rodents. Despite an overall fast turnover of TF binding locations between species, we identified thousands of TF regions of highly constrained TF binding intensity. Although individual mutations in bound sequence motifs can influence TF binding, most binding differences occur in the absence of nearby sequence variations. Instead, combinatorial binding was found to be significant for genetic and evolutionary stability; cobound TFs tend to disappear in concert and were sensitive to genetic knockout of partner TFs. The large, qualitative differences in genomic regions bound between closely related mammals, when contrasted with the smaller, quantitative TF binding differences among Drosophila species, illustrate how genome structure and population genetics together shape regulatory evolution.


Asunto(s)
Evolución Molecular , Ratones/clasificación , Ratones/genética , Factores de Transcripción/genética , Animales , Drosophila/genética , Hígado/metabolismo , Ratones/metabolismo , Ratones Endogámicos , Ratones Noqueados , Ratas/genética , Factores de Transcripción/metabolismo
18.
Clin Cancer Res ; 19(11): 2810-6, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23549874

RESUMEN

Inflammatory signals from the surrounding microenvironment play important roles in tumor promotion. Key inflammatory mediators and pathways that induce and sustain tumorigenesis have recently been identified in many different cancers. Hepatocellular carcinoma is a paradigm for inflammation-induced cancer, as it most frequently develops in the setting of chronic hepatitis, consecutive cellular damage, and compensatory regeneration. Recent studies revealed that liver damage-mediated inflammation and carcinogenesis are triggered by a complex cross-talk between NF-κB, c-jun-NH2-kinase, and STAT3 signaling pathways. Molecular dissection of the mechanisms involved in the interplay between these pathways identified promising new targets for therapeutic intervention. Targeting different components of the signaling cascades may provide efficient means for blocking the apparently irreversible sequence of events initiated by chronic liver inflammation and culminating in liver cancer.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Transducción de Señal , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Humanos , Inflamación/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Investigación Biomédica Traslacional
19.
Cancer Cell ; 21(6): 738-50, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22698400

RESUMEN

The tumor suppressor cylindromatosis (CYLD) inhibits the NFκB and mitogen-activated protein kinase (MAPK) activation pathways by deubiquitinating upstream regulatory factors. Here we show that liver-specific disruption of CYLD triggers hepatocyte cell death in the periportal area via spontaneous and chronic activation of TGF-ß activated kinase 1 (TAK1) and c-Jun N-terminal kinase (JNK). This is followed by hepatic stellate cell and Kupffer cell activation, which promotes progressive fibrosis, inflammation, tumor necrosis factor (TNF) production, and expansion of hepatocyte apoptosis toward the central veins. At later stages, compensatory proliferation results in the development of cancer foci featuring re-expression of oncofetal hepatic and stem cell-specific genes. The results demonstrate that, in the liver, CYLD acts as an important regulator of hepatocyte homeostasis, protecting cells from spontaneous apoptosis by preventing uncontrolled TAK1 and JNK activation.


Asunto(s)
Apoptosis/genética , Cisteína Endopeptidasas/genética , Hepatocitos/metabolismo , Hígado/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Hibridación Genómica Comparativa , Cisteína Endopeptidasas/metabolismo , Enzima Desubiquitinante CYLD , Activación Enzimática/efectos de los fármacos , Fibrosis/genética , Fibrosis/metabolismo , Perfilación de la Expresión Génica , Hepatocitos/patología , Inmunohistoquímica , Inflamación/genética , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
20.
Proc Natl Acad Sci U S A ; 109(8): 2995-3000, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22323595

RESUMEN

Nucleotide excision repair (NER) defects are associated with cancer, developmental disorders and neurodegeneration. However, with the exception of cancer, the links between defects in NER and developmental abnormalities are not well understood. Here, we show that the ERCC1-XPF NER endonuclease assembles on active promoters in vivo and facilitates chromatin modifications for transcription during mammalian development. We find that Ercc1(-/-) mice demonstrate striking physiological, metabolic and gene expression parallels with Taf10(-/-) animals carrying a liver-specific transcription factor II D (TFIID) defect in transcription initiation. Promoter occupancy studies combined with expression profiling in the liver and in vitro differentiation cell assays reveal that ERCC1-XPF interacts with TFIID and assembles with POL II and the basal transcription machinery on promoters in vivo. Whereas ERCC1-XPF is required for the initial activation of genes associated with growth, it is dispensable for ongoing transcription. Recruitment of ERCC1-XPF on promoters is accompanied by promoter-proximal DNA demethylation and histone marks associated with active hepatic transcription. Collectively, the data unveil a role of ERCC1/XPF endonuclease in transcription initiation establishing its causal contribution to NER developmental disorders.


Asunto(s)
Reparación del ADN/genética , Crecimiento y Desarrollo/genética , Progeria/genética , Transcripción Genética , Adipogénesis/genética , Animales , Animales Recién Nacidos , Metilación de ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Endonucleasas/deficiencia , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Histonas/metabolismo , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Hígado/patología , Ratones , Especificidad de Órganos , Progeria/enzimología , Progeria/patología , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Procesamiento Proteico-Postraduccional/genética , Factor de Transcripción TFIID/metabolismo , Transcriptoma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...