Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(3): 564-580.e19, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30580964

RESUMEN

There are still gaps in our understanding of the complex processes by which p53 suppresses tumorigenesis. Here we describe a novel role for p53 in suppressing the mevalonate pathway, which is responsible for biosynthesis of cholesterol and nonsterol isoprenoids. p53 blocks activation of SREBP-2, the master transcriptional regulator of this pathway, by transcriptionally inducing the ABCA1 cholesterol transporter gene. A mouse model of liver cancer reveals that downregulation of mevalonate pathway gene expression by p53 occurs in premalignant hepatocytes, when p53 is needed to actively suppress tumorigenesis. Furthermore, pharmacological or RNAi inhibition of the mevalonate pathway restricts the development of murine hepatocellular carcinomas driven by p53 loss. Like p53 loss, ablation of ABCA1 promotes murine liver tumorigenesis and is associated with increased SREBP-2 maturation. Our findings demonstrate that repression of the mevalonate pathway is a crucial component of p53-mediated liver tumor suppression and outline the mechanism by which this occurs.


Asunto(s)
Ácido Mevalónico/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Línea Celular , Colesterol/metabolismo , Femenino , Genes Supresores de Tumor , Células HCT116 , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/genética , Regiones Promotoras Genéticas , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Terpenos/metabolismo
2.
Cell ; 161(7): 1539-1552, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26091037

RESUMEN

The adenomatous polyposis coli (APC) tumor suppressor is mutated in the vast majority of human colorectal cancers (CRC) and leads to deregulated Wnt signaling. To determine whether Apc disruption is required for tumor maintenance, we developed a mouse model of CRC whereby Apc can be conditionally suppressed using a doxycycline-regulated shRNA. Apc suppression produces adenomas in both the small intestine and colon that, in the presence of Kras and p53 mutations, can progress to invasive carcinoma. In established tumors, Apc restoration drives rapid and widespread tumor-cell differentiation and sustained regression without relapse. Tumor regression is accompanied by the re-establishment of normal crypt-villus homeostasis, such that once aberrantly proliferating cells reacquire self-renewal and multi-lineage differentiation capability. Our study reveals that CRC cells can revert to functioning normal cells given appropriate signals and provide compelling in vivo validation of the Wnt pathway as a therapeutic target for treatment of CRC.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Intestino Grueso/patología , Intestino Delgado/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proliferación Celular , Neoplasias Colorrectales/patología , Doxiciclina/administración & dosificación , Genes p53 , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Intestino Grueso/metabolismo , Intestino Delgado/metabolismo , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas p21(ras)/genética , Interferencia de ARN , Vía de Señalización Wnt
3.
Cell ; 158(3): 579-92, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083869

RESUMEN

The p53 tumor suppressor coordinates a series of antiproliferative responses that restrict the expansion of malignant cells, and as a consequence, p53 is lost or mutated in the majority of human cancers. Here, we show that p53 restricts expression of the stem and progenitor-cell-associated protein nestin in an Sp1/3 transcription-factor-dependent manner and that Nestin is required for tumor initiation in vivo. Moreover, loss of p53 facilitates dedifferentiation of mature hepatocytes into nestin-positive progenitor-like cells, which are poised to differentiate into hepatocellular carcinomas (HCCs) or cholangiocarcinomas (CCs) in response to lineage-specific mutations that target Wnt and Notch signaling, respectively. Many human HCCs and CCs show elevated nestin expression, which correlates with p53 loss of function and is associated with decreased patient survival. Therefore, transcriptional repression of Nestin by p53 restricts cellular plasticity and tumorigenesis in liver cancer.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Nestina/metabolismo , Animales , Carcinoma Hepatocelular/patología , Transformación Celular Neoplásica , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones , Pronóstico , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Cell ; 157(2): 382-394, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725405

RESUMEN

Missense mutations in the p53 tumor suppressor inactivate its antiproliferative properties but can also promote metastasis through a gain-of-function activity. We show that sustained expression of mutant p53 is required to maintain the prometastatic phenotype of a murine model of pancreatic cancer, a highly metastatic disease that frequently displays p53 mutations. Transcriptional profiling and functional screening identified the platelet-derived growth factor receptor b (PDGFRb) as both necessary and sufficient to mediate these effects. Mutant p53 induced PDGFRb through a cell-autonomous mechanism involving inhibition of a p73/NF-Y complex that represses PDGFRb expression in p53-deficient, noninvasive cells. Blocking PDGFRb signaling by RNA interference or by small molecule inhibitors prevented pancreatic cancer cell invasion in vitro and metastasis formation in vivo. Finally, high PDGFRb expression correlates with poor disease-free survival in pancreatic, colon, and ovarian cancer patients, implicating PDGFRb as a prognostic marker and possible target for attenuating metastasis in p53 mutant tumors.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Carcinoma Ductal Pancreático/patología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteína p53 Supresora de Tumor/genética
5.
Cell ; 153(2): 449-60, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23562644

RESUMEN

The p53 tumor suppressor can restrict malignant transformation by triggering cell-autonomous programs of cell-cycle arrest or apoptosis. p53 also promotes cellular senescence, a tumor-suppressive program that involves stable cell-cycle arrest and secretion of factors that modify the tissue microenvironment. In the presence of chronic liver damage, we show that ablation of a p53-dependent senescence program in hepatic stellate cells increases liver fibrosis and cirrhosis associated with reduced survival and enhances the transformation of adjacent epithelial cells into hepatocellular carcinoma. p53-expressing senescent stellate cells release factors that skew macrophage polarization toward a tumor-inhibiting M1-state capable of attacking senescent cells in culture, whereas proliferating p53-deficient stellate cells secrete factors that stimulate polarization of macrophages into a tumor-promoting M2-state and enhance the proliferation of premalignant cells. Hence, p53 can act non-cell autonomously to suppress tumorigenesis by promoting an antitumor microenvironment, in part, through secreted factors that modulate macrophage function.


Asunto(s)
Transformación Celular Neoplásica , Senescencia Celular , Células Estrelladas Hepáticas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Microambiente Celular , Fibrosis/patología , Células Estrelladas Hepáticas/citología , Humanos , Inflamación/metabolismo , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Hígado/citología , Hígado/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , FN-kappa B
7.
Proc Natl Acad Sci U S A ; 119(17): e2110557119, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35442775

RESUMEN

Anticancer drug development campaigns often fail due to an incomplete understanding of the therapeutic index differentiating the efficacy of the agent against the cancer and its on-target toxicities to the host. To address this issue, we established a versatile preclinical platform in which genetically defined cancers are produced using somatic tissue engineering in transgenic mice harboring a doxycycline-inducible short hairpin RNA against the target of interest. In this system, target inhibition is achieved by the addition of doxycycline, enabling simultaneous assessment of efficacy and toxicity in the same animal. As proof of concept, we focused on CDK9­a cancer target whose clinical development has been hampered by compounds with poorly understood target specificity and unacceptable toxicities. We systematically compared phenotypes produced by genetic Cdk9 inhibition to those achieved using a recently developed highly specific small molecule CDK9 inhibitor and found that both perturbations led to robust antitumor responses. Remarkably, nontoxic levels of CDK9 inhibition could achieve significant treatment efficacy, and dose-dependent toxicities produced by prolonged CDK9 suppression were largely reversible upon Cdk9 restoration or drug withdrawal. Overall, these results establish a versatile in vivo target validation platform that can be employed for rapid triaging of therapeutic targets and lend support to efforts aimed at advancing CDK9 inhibitors for cancer therapy.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Interferencia de ARN
8.
Gut ; 71(8): 1613-1628, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34509979

RESUMEN

OBJECTIVE: Large-scale genome sequencing efforts of human tumours identified epigenetic modifiers as one of the most frequently mutated gene class in human cancer. However, how these mutations drive tumour development and tumour progression are largely unknown. Here, we investigated the function of the histone demethylase KDM6A in gastrointestinal cancers, such as liver cancer and pancreatic cancer. DESIGN: Genetic alterations as well as expression analyses of KDM6A were performed in patients with liver cancer. Genetic mouse models of liver and pancreatic cancer coupled with Kdm6a-deficiency were investigated, transcriptomic and epigenetic profiling was performed, and in vivo and in vitro drug treatments were conducted. RESULTS: KDM6A expression was lost in 30% of patients with liver cancer. Kdm6a deletion significantly accelerated tumour development in murine liver and pancreatic cancer models. Kdm6a-deficient tumours showed hyperactivation of mTORC1 signalling, whereas endogenous Kdm6a re-expression by inducible RNA-interference in established Kdm6a-deficient tumours diminished mTORC1 activity resulting in attenuated tumour progression. Genome-wide transcriptional and epigenetic profiling revealed direct binding of Kdm6a to crucial negative regulators of mTORC1, such as Deptor, and subsequent transcriptional activation by epigenetic remodelling. Moreover, in vitro and in vivo genetic epistasis experiments illustrated a crucial function of Deptor and mTORC1 in Kdm6a-dependent tumour suppression. Importantly, KDM6A expression in human tumours correlates with mTORC1 activity and KDM6A-deficient tumours exhibit increased sensitivity to mTORC1 inhibition. CONCLUSION: KDM6A is an important tumour suppressor in gastrointestinal cancers and acts as an epigenetic toggle for mTORC1 signalling. Patients with KDM6A-deficient tumours could benefit of targeted therapy focusing on mTORC1 inhibition.


Asunto(s)
Histona Demetilasas/metabolismo , Neoplasias Hepáticas , Neoplasias Pancreáticas , Animales , Epigénesis Genética , Histona Demetilasas/genética , Histonas/genética , Neoplasias Hepáticas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas
9.
Nat Methods ; 16(1): 51-54, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559432

RESUMEN

CRISPR-Cas9-based combinatorial perturbation approaches for orthogonal knockout and gene activation have been impeded by complex vector designs and co-delivery of multiple constructs. Here, we demonstrate that catalytically active CRISPR-Cas12a fused to a transcriptional-activator domain enables flexible switching between genome editing and transcriptional activation by altering guide length. By leveraging Cas12a-mediated CRISPR-RNA array processing, we illustrate that Cas12a-VPR enables simplified multiplexed knockout and transcriptional activation in vitro and in vivo.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Activación Transcripcional , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones
10.
Genes Dev ; 28(16): 1800-14, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25128497

RESUMEN

One-year survival rates for newly diagnosed hepatocellular carcinoma (HCC) are <50%, and unresectable HCC carries a dismal prognosis owing to its aggressiveness and the undruggable nature of its main genetic drivers. By screening a custom library of shRNAs directed toward known drug targets in a genetically defined Myc-driven HCC model, we identified cyclin-dependent kinase 9 (Cdk9) as required for disease maintenance. Pharmacological or shRNA-mediated CDK9 inhibition led to robust anti-tumor effects that correlated with MYC expression levels and depended on the role that both CDK9 and MYC exert in transcription elongation. Our results establish CDK9 inhibition as a therapeutic strategy for MYC-overexpressing liver tumors and highlight the relevance of transcription elongation in the addiction of cancer cells to MYC.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Quinasa 9 Dependiente de la Ciclina/metabolismo , Neoplasias Hepáticas/enzimología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Elongación de la Transcripción Genética/fisiología , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Femenino , Expresión Génica , Biblioteca de Genes , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
11.
Proc Natl Acad Sci U S A ; 114(50): 13076-13084, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29162699

RESUMEN

A segmental deletion resulting in DNAJB1-PRKACA gene fusion is now recognized as the signature genetic event of fibrolamellar hepatocellular carcinoma (FL-HCC), a rare but lethal liver cancer that primarily affects adolescents and young adults. Here we implement CRISPR-Cas9 genome editing and transposon-mediated somatic gene transfer to demonstrate that expression of either the endogenous fusion protein or a chimeric cDNA leads to the formation of indolent liver tumors in mice that closely resemble human FL-HCC. Notably, overexpression of the wild-type PRKACA was unable to fully recapitulate the oncogenic activity of DNAJB1-PRKACA, implying that FL-HCC does not simply result from enhanced PRKACA expression. Tumorigenesis was significantly enhanced by genetic activation of ß-catenin, an observation supported by evidence of recurrent Wnt pathway mutations in human FL-HCC, as well as treatment with the hepatotoxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine, which causes tissue injury, inflammation, and fibrosis. Our study validates the DNAJB1-PRKACA fusion kinase as an oncogenic driver and candidate drug target for FL-HCC, and establishes a practical model for preclinical studies to identify strategies to treat this disease.


Asunto(s)
Carcinoma Hepatocelular/genética , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Proteínas del Choque Térmico HSP40/genética , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas/genética , Regeneración Hepática/genética , Hígado/fisiología , Proteínas de Fusión Oncogénica/genética , beta Catenina/genética , Adulto , Animales , Secuencia de Bases , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Cromosomas Humanos Par 19/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Estudios de Cohortes , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Piridinas/toxicidad , Eliminación de Secuencia/genética , Adulto Joven
12.
Nat Mater ; 17(4): 361-368, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29403054

RESUMEN

Development of targeted nanoparticle drug carriers often requires complex synthetic schemes involving both supramolecular self-assembly and chemical modification. These processes are generally difficult to predict, execute, and control. We describe herein a targeted drug delivery system that is accurately and quantitatively predicted to self-assemble into nanoparticles based on the molecular structures of precursor molecules, which are the drugs themselves. The drugs assemble with the aid of sulfated indocyanines into particles with ultrahigh drug loadings of up to 90%. We devised quantitative structure-nanoparticle assembly prediction (QSNAP) models to identify and validate electrotopological molecular descriptors as highly predictive indicators of nano-assembly and nanoparticle size. The resulting nanoparticles selectively targeted kinase inhibitors to caveolin-1-expressing human colon cancer and autochthonous liver cancer models to yield striking therapeutic effects while avoiding pERK inhibition in healthy skin. This finding enables the computational design of nanomedicines based on quantitative models for drug payload selection.


Asunto(s)
Portadores de Fármacos/química , Nanomedicina/métodos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Endocitosis , Indoles/química , Ratones , Nanopartículas/química , Tamaño de la Partícula , Distribución Tisular
13.
Genome Med ; 16(1): 83, 2024 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886830

RESUMEN

BACKGROUND: Somatic copy number alterations are a hallmark of cancer that offer unique opportunities for therapeutic exploitation. Here, we focused on the identification of specific vulnerabilities for tumors harboring chromosome 8p deletions. METHODS: We developed and applied an integrative analysis of The Cancer Genome Atlas (TCGA), the Cancer Dependency Map (DepMap), and the Cancer Cell Line Encyclopedia to identify chromosome 8p-specific vulnerabilities. We employ orthogonal gene targeting strategies, both in vitro and in vivo, including short hairpin RNA-mediated gene knockdown and CRISPR/Cas9-mediated gene knockout to validate vulnerabilities. RESULTS: We identified SLC25A28 (also known as MFRN2), as a specific vulnerability for tumors harboring chromosome 8p deletions. We demonstrate that vulnerability towards MFRN2 loss is dictated by the expression of its paralog, SLC25A37 (also known as MFRN1), which resides on chromosome 8p. In line with their function as mitochondrial iron transporters, MFRN1/2 paralog protein deficiency profoundly impaired mitochondrial respiration, induced global depletion of iron-sulfur cluster proteins, and resulted in DNA-damage and cell death. MFRN2 depletion in MFRN1-deficient tumors led to impaired growth and even tumor eradication in preclinical mouse xenograft experiments, highlighting its therapeutic potential. CONCLUSIONS: Our data reveal MFRN2 as a therapeutic target of chromosome 8p deleted cancers and nominate MFNR1 as the complimentary biomarker for MFRN2-directed therapies.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 8 , Neoplasias , Humanos , Cromosomas Humanos Par 8/genética , Animales , Ratones , Neoplasias/genética , Línea Celular Tumoral , Mutaciones Letales Sintéticas , Mitocondrias/metabolismo , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Regulación Neoplásica de la Expresión Génica , Variaciones en el Número de Copia de ADN
14.
Eur J Immunol ; 42(12): 3369-80, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23001948

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) presenting with a micropapillary growth pattern is frequently associated with a prominent neutrophil infiltration into the tumor. The relevance of neutrophil infiltrates for tumor progression, however, is still debated. To gain insight into the role of polymorphonuclear neutrophils (PMNs) in PDAC, we assessed their effect on pancreatic tumor cells grown in vitro as monolayers. Time-lapse video microscopy showed a PMN-induced dyshesion of the tumor cells, and subsequent experiments revealed that this dyshesion was due to PMN elastase-mediated degradation of E-cadherin, an adhesion molecule that mediates the intercellular contact of the tumor cells. E-cadherin degradation by elastase or--(for comparison) down-modulation by specific siRNA, significantly increased the migratory capacity of the pancreatic tumor cells, leading to the hypothesis that PMNs could contribute to the invasive tumor growth. To address this issue, biopsies of patients with PDAC (n = 112) were analyzed. We found that E-cadherin expression correlated negatively with PMN infiltration, compatible with the notion that E-cadherin is cleaved by PMN-derived elastase, which in turn could result in the dispersal of the tumor cells, enhanced migratory capacity and thus invasive tumor growth.


Asunto(s)
Cadherinas/inmunología , Carcinoma Ductal Pancreático/inmunología , Elastasa de Leucocito/inmunología , Proteínas de Neoplasias/inmunología , Neutrófilos/inmunología , Neoplasias Pancreáticas/inmunología , Proteolisis , Adulto , Anciano , Anciano de 80 o más Años , Cadherinas/biosíntesis , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Adhesión Celular/inmunología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Elastasa de Leucocito/metabolismo , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Infiltración Neutrófila/inmunología , Neutrófilos/enzimología , Neutrófilos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología
15.
Gut ; 61(8): 1209-18, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22287599

RESUMEN

BACKGROUND: At present hepatocyte transplantation is a promising option for cellular therapy of end-stage liver diseases. However, the underlying molecular mechanisms need to be better defined in order to translate this technique into clinical use. This study investigated the cursiv relevance of hepatocyte growth factor (HGF)/c-Met signalling for hepatocyte repopulation after transplantion. METHODS: Wild-type mice (c-Met(loxP/loxP)) and hepatocyte-specific conditional c-Met (HGF receptor) knockout (c-Met(Δhepa)) mice were used as donors and recipients for hepatocyte transplantation. RESULTS: Transplantation experiments revealed two major findings. First it was demonstrated that c-Met is indispensable in donor cells, as c-Met(Δhepa) cells did not repopulate recipient livers after transplantation. Second, genetic deletion of c-Met in recipient hepatocytes resulted in enhanced expansion of unmodified donor cells in host livers (up to 250-fold after 12 weeks). The relevant mechanisms for this observation in c-Met(Δhepa) host hepatocytes could be defined. c-Met(Δhepa) hepatocytes showed enhanced apoptosis, reduced cellular proliferation and a lack of AKT-kinase and STAT3 activation. In addition, tissue remodelling was changed in c-Met(Δhepa) recipient livers. Therefore, the lack of pro-proliferative transcription factors, increased apoptosis and changes in matrix-remodelling inhibit host cell proliferation in c-Met(Δhepa) recipient livers and thus favour repopulation of transplanted hepatocytes. Therapeutically liver repopulation could be increased through adenoviral expression of NK-4--an inhibitor of HGF signalling--in host hepatocytes. CONCLUSION: HGF/c-Met plays a crucial role in host and donor cells of the liver for the cursiv selection of transplanted hepatocytes. Modulating HGF-dependent signalling seems a promising therapeutic option to favour expansion of transplanted hepatocytes.


Asunto(s)
Regulación de la Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Hepatocitos/trasplante , Regeneración Hepática , Trasplante de Hígado/métodos , ARN Mensajero/genética , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Apoptosis , Western Blotting , Comunicación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor de Crecimiento de Hepatocito/biosíntesis , Hepatocitos/citología , Etiquetado Corte-Fin in Situ , Fallo Hepático/genética , Fallo Hepático/metabolismo , Fallo Hepático/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Transducción de Señal
16.
Elife ; 122023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37261974

RESUMEN

Mutations in genes encoding components of chromatin modifying and remodeling complexes are among the most frequently observed somatic events in human cancers. For example, missense and nonsense mutations targeting the mixed lineage leukemia family member 3 (MLL3, encoded by KMT2C) histone methyltransferase occur in a range of solid tumors, and heterozygous deletions encompassing KMT2C occur in a subset of aggressive leukemias. Although MLL3 loss can promote tumorigenesis in mice, the molecular targets and biological processes by which MLL3 suppresses tumorigenesis remain poorly characterized. Here, we combined genetic, epigenomic, and animal modeling approaches to demonstrate that one of the mechanisms by which MLL3 links chromatin remodeling to tumor suppression is by co-activating the Cdkn2a tumor suppressor locus. Disruption of Kmt2c cooperates with Myc overexpression in the development of murine hepatocellular carcinoma (HCC), in which MLL3 binding to the Cdkn2a locus is blunted, resulting in reduced H3K4 methylation and low expression levels of the locus-encoded tumor suppressors p16/Ink4a and p19/Arf. Conversely, elevated KMT2C expression increases its binding to the CDKN2A locus and co-activates gene transcription. Endogenous Kmt2c restoration reverses these chromatin and transcriptional effects and triggers Ink4a/Arf-dependent apoptosis. Underscoring the human relevance of this epistasis, we found that genomic alterations in KMT2C and CDKN2A were associated with similar transcriptional profiles in human HCC samples. These results collectively point to a new mechanism for disrupting CDKN2A activity during cancer development and, in doing so, link MLL3 to an established tumor suppressor network.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animales , Ratones , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteína p14ARF Supresora de Tumor/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Cromatina , Carcinogénesis
17.
Am J Pathol ; 176(5): 2236-46, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20382701

RESUMEN

The 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) model leads to chronic cholestatic liver injury and therefore resembles human diseases such as sclerosing cholangitis and forms of metabolic liver diseases. The role of the interleukin-6/glycoprotein 130 (gp130) system in this context is still undefined. Therefore, conditional gp130 knockout and knockin mice were used to achieve hepatocyte-specific deletions of gp130 (gp130(Deltahepa)), gp130-dependent ras (gp130(DeltahepaRas)), and signal transducer and activator of transcription (STAT) (gp130(DeltahepaSTAT)) activation. These mice were treated with a DDC-containing diet and analyzed over time. Mice deficient in hepatic gp130 and STAT signaling showed increased and earlier mortality than wild-type and gp130(DeltahepaRas) animals. Over time, significantly more apoptosis and cholestasis became evident in gp130(Deltahepa) and gp130(DeltahepaSTAT) mice. These mice also displayed increased tumor necrosis factor-alpha expression, a diminished acute-phase response (lack of STAT3 and serum amyloid A activation), and enhanced immune cell infiltration in the liver. These were associated with stronger periportal oval cell activation. In addition, DDC treatment in gp130(Deltahepa) and gp130(DeltahepaSTAT) mice resulted in significantly stronger hepatic stellate cell activation. Long-term analysis revealed the development of severe liver fibrosis in gp130(Deltahepa) and gp130(DeltahepaSTAT) animals, as evidenced by increased collagen accumulation. Here we demonstrate that gp130/STAT signaling in hepatocytes provides protection in a cholestatic hepatitis mouse model. STAT3-dependent signaling pathways in hepatocytes protect from apoptosis and tissue injury, which subsequently reduce oval cell activation and prevent fibrosis progression.


Asunto(s)
Colangitis Esclerosante/patología , Receptor gp130 de Citocinas/metabolismo , Fibrosis/metabolismo , Hepatocitos/citología , Hígado/lesiones , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Alelos , Animales , Apoptosis , Colestasis/patología , Progresión de la Enfermedad , Hígado/patología , Masculino , Ratones , Ratones Transgénicos
18.
Hepatology ; 51(2): 463-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19918973

RESUMEN

UNLABELLED: A deregulated cytokine balance is involved in triggering the sequence from steatosis to nonalcoholic steatohepatitis, ultimately leading to liver fibrosis and cancer. To better define the role of proinflammatory interleukin-6 (IL-6)-type cytokines in hepatocytes we investigated the role of IL-6 and its shared receptor, glycoprotein 130 (gp130), in a mouse model of steatohepatitis. IL-6(-/-) mice were fed a choline-deficient, ethionine-supplemented (CDE) diet. Conditional gp130 knockout and knockin mice were used to achieve hepatocyte-specific deletion of gp130 (gp130(Deltahepa)), gp130-dependent rat sarcoma (Ras)-(gp130(DeltahepaRas)), and signal transducers and activators of transcription (STAT)-(gp130(DeltahepaSTAT)) activation. CDE-treated IL-6(-/-) mice showed a significant hepatic steatosis at 2 weeks after feeding. The mice rapidly developed elevated fasting blood glucose, insulin serum levels, and transaminases. To better define IL-6-dependent intracellular pathways, specifically in hepatocytes, we next treated gp130(Deltahepa) mice with a CDE diet. These animals also developed a marked steatosis with hyperglycemia and displayed elevated insulin serum levels. Additionally, gp130(Deltahepa) animals showed an imbalanced inflammatory response with increased hepatic tumor necrosis factor-alpha and decreased adiponectin messenger RNA levels. Dissecting the hepatocyte-specific gp130-dependent pathways revealed a similar disease phenotype in gp130(DeltahepaSTAT) mice, whereas gp130(DeltahepaRas) animals were protected. In CDE-treated mice lack of gp130-STAT3 signaling was associated with immune-cell-infiltration, jun kinase-activation, a blunted acute-phase-response, and elevated transaminases. Furthermore, gp130(Deltahepa) and gp130(DeltahepaSTAT) mice showed beginning signs of liver fibrosis compared to gp130(DeltahepaRas) mice and controls. CONCLUSION: During CDE treatment mice lacking IL-6 and gp130-STAT signaling in hepatocytes are prone to hepatic metabolic changes and inflammation. This ultimately leads to progressive steatohepatitis with signs of liver remodeling. Thus, the presented model allows one to further dissect the role of IL-6/gp130-type signaling in hepatocytes during fatty liver degeneration to define new therapeutic targets in metabolic liver diseases.


Asunto(s)
Factor de Transcripción Activador 3/fisiología , Receptor gp130 de Citocinas/fisiología , Hígado Graso/etiología , Hepatitis/etiología , Hepatocitos , Transducción de Señal/fisiología , Animales , Causalidad , Ratones
19.
Liver Transpl ; 16(1): 23-32, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20035522

RESUMEN

Hepatocyte transplantation (HT) is still restricted by the limited amount of transplantable cells. Therefore, a better understanding of the mechanisms involved in cellular engraftment, proliferation, and in vivo selection is important. Here we aimed to evaluate the role of the interleukin 6 (IL-6)/glycoprotein 130 (gp130) system for liver repopulation. Mice carrying a conditional hepatocyte-specific deletion of the common IL-6 signal transducer gp130 (gp130(Deltahepa)) were used for HT. First, we compared bone marrow transplantation (BMT), partial hepatectomy (PH), and retrorsine treatment of recipient mice to optimize the in vivo selection of transplanted hepatocytes. BMT combined with PH was sufficient to induce a 30-fold increase in the number of transplanted donor hepatocytes, whereas additional retrorsine pretreatment led to an up to 40-fold increase. Next, the influence of gp130 signaling in hepatocytes on cell selection was evaluated. Wild-type (WT) hepatocytes repopulated WT recipients at the same rate as gp130(Deltahepa) cells. In contrast, liver repopulation by transplanted cells was enhanced in gp130(Deltahepa) recipient mice. This was associated with higher proliferation of donor hepatocytes and enhanced apoptosis in gp130(Deltahepa) recipient livers. Additionally, the acute phase response was strongly induced after HT in WT recipients but blunted in gp130(Deltahepa) recipients. As a result, significantly more liver remodeling, evidenced by stronger hepatic stellate cell activation and collagen accumulation, was found in gp130(Deltahepa) mice after HT. In conclusion, the HT model established here can be efficiently applied to investigate cell-specific mechanisms in liver repopulation. Moreover, we have shown that gp130-dependent pathways in host hepatocytes are important for controlling liver repopulation.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Hepatocitos/trasplante , Hígado/metabolismo , Reacción de Fase Aguda/metabolismo , Animales , Colágeno/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Ratones , Ratones Transgénicos , Transducción de Señal
20.
Cell Rep ; 30(10): 3280-3295.e6, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32160537

RESUMEN

Genome editing technologies have transformed our ability to engineer desired genomic changes within living systems. However, detecting precise genomic modifications often requires sophisticated, expensive, and time-consuming experimental approaches. Here, we describe DTECT (Dinucleotide signaTurE CapTure), a rapid and versatile detection method that relies on the capture of targeted dinucleotide signatures resulting from the digestion of genomic DNA amplicons by the type IIS restriction enzyme AcuI. DTECT enables the accurate quantification of marker-free precision genome editing events introduced by CRISPR-dependent homology-directed repair, base editing, or prime editing in various biological systems, such as mammalian cell lines, organoids, and tissues. Furthermore, DTECT allows the identification of oncogenic mutations in cancer mouse models, patient-derived xenografts, and human cancer patient samples. The ease, speed, and cost efficiency by which DTECT identifies genomic signatures should facilitate the generation of marker-free cellular and animal models of human disease and expedite the detection of human pathogenic variants.


Asunto(s)
Edición Génica , Variación Genética , Genómica , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Secuencia de Bases , ADN/genética , Modelos Animales de Enfermedad , Sitios Genéticos , Marcadores Genéticos , Genotipo , Células HEK293 , Humanos , Ratones , Mutación/genética , Células 3T3 NIH , Neoplasias/genética , Nucleótidos/genética , Oncogenes , Reparación del ADN por Recombinación/genética , Mapeo Restrictivo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA