Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Metab ; 5(8): 1303-1318, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37580540

RESUMEN

The genomic landscape of colorectal cancer (CRC) is shaped by inactivating mutations in tumour suppressors such as APC, and oncogenic mutations such as mutant KRAS. Here we used genetically engineered mouse models, and multimodal mass spectrometry-based metabolomics to study the impact of common genetic drivers of CRC on the metabolic landscape of the intestine. We show that untargeted metabolic profiling can be applied to stratify intestinal tissues according to underlying genetic alterations, and use mass spectrometry imaging to identify tumour, stromal and normal adjacent tissues. By identifying ions that drive variation between normal and transformed tissues, we found dysregulation of the methionine cycle to be a hallmark of APC-deficient CRC. Loss of Apc in the mouse intestine was found to be sufficient to drive expression of one of its enzymes, adenosylhomocysteinase (AHCY), which was also found to be transcriptionally upregulated in human CRC. Targeting of AHCY function impaired growth of APC-deficient organoids in vitro, and prevented the characteristic hyperproliferative/crypt progenitor phenotype driven by acute deletion of Apc in vivo, even in the context of mutant Kras. Finally, pharmacological inhibition of AHCY reduced intestinal tumour burden in ApcMin/+ mice indicating its potential as a metabolic drug target in CRC.


Asunto(s)
Neoplasias Colorrectales , Animales , Humanos , Ratones , Adenosilhomocisteinasa/genética , Adenosilhomocisteinasa/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Metabolómica , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética
2.
Sci Adv ; 7(47): eabj0512, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34788095

RESUMEN

Somatic mutations in APC or CTNNB1 genes lead to aberrant Wnt signaling and colorectal cancer (CRC) initiation and progression via-catenin­T cell factor/lymphoid enhancer binding factor TCF/LEF transcription factors. We found that Lef1 was expressed exclusively in Apc-mutant, Wnt ligand­independent tumors, but not in ligand-dependent, serrated tumors. To analyze Lef1 function in tumor development, we conditionally deleted Lef1 in intestinal stem cells of Apcfl/fl mice or broadly from the entire intestinal epithelium of Apcfl/fl or ApcMin/+ mice. Loss of Lef1 markedly increased tumor initiation and tumor cell proliferation, reduced the expression of several Wnt antagonists, and increased Myc proto-oncogene expression and formation of ectopic crypts in Apc-mutant adenomas. Our results uncover a previously unknown negative feedback mechanism in CRC, in which ectopic Lef1 expression suppresses intestinal tumorigenesis by restricting adenoma cell dedifferentiation to a crypt-progenitor phenotype and by reducing the formation of cancer stem cell niches.

3.
Nat Commun ; 12(1): 3464, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103493

RESUMEN

Right-sided (proximal) colorectal cancer (CRC) has a poor prognosis and a distinct mutational profile, characterized by oncogenic BRAF mutations and aberrations in mismatch repair and TGFß signalling. Here, we describe a mouse model of right-sided colon cancer driven by oncogenic BRAF and loss of epithelial TGFß-receptor signalling. The proximal colonic tumours that develop in this model exhibit a foetal-like progenitor phenotype (Ly6a/Sca1+) and, importantly, lack expression of Lgr5 and its associated intestinal stem cell signature. These features are recapitulated in human BRAF-mutant, right-sided CRCs and represent fundamental differences between left- and right-sided disease. Microbial-driven inflammation supports the initiation and progression of these tumours with foetal-like characteristics, consistent with their predilection for the microbe-rich right colon and their antibiotic sensitivity. While MAPK-pathway activating mutations drive this foetal-like signature via ERK-dependent activation of the transcriptional coactivator YAP, the same foetal-like transcriptional programs are also initiated by inflammation in a MAPK-independent manner. Importantly, in both contexts, epithelial TGFß-receptor signalling is instrumental in suppressing the tumorigenic potential of these foetal-like progenitor cells.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias del Colon/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinogénesis/patología , Diferenciación Celular , Supervivencia Celular , Colon/patología , Neoplasias del Colon/genética , Células Epiteliales/metabolismo , Feto/patología , Inflamación/patología , Estimación de Kaplan-Meier , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Mutación , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Proteínas Señalizadoras YAP
4.
Nature ; 594(7863): 430-435, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34079124

RESUMEN

The tumour suppressor APC is the most commonly mutated gene in colorectal cancer. Loss of Apc in intestinal stem cells drives the formation of adenomas in mice via increased WNT signalling1, but reduced secretion of WNT ligands increases the ability of Apc-mutant intestinal stem cells to colonize a crypt (known as fixation)2. Here we investigated how Apc-mutant cells gain a clonal advantage over wild-type counterparts to achieve fixation. We found that Apc-mutant cells are enriched for transcripts that encode several secreted WNT antagonists, with Notum being the most highly expressed. Conditioned medium from Apc-mutant cells suppressed the growth of wild-type organoids in a NOTUM-dependent manner. Furthermore, NOTUM-secreting Apc-mutant clones actively inhibited the proliferation of surrounding wild-type crypt cells and drove their differentiation, thereby outcompeting crypt cells from the niche. Genetic or pharmacological inhibition of NOTUM abrogated the ability of Apc-mutant cells to expand and form intestinal adenomas. We identify NOTUM as a key mediator during the early stages of mutation fixation that can be targeted to restore wild-type cell competitiveness and provide preventative strategies for people at a high risk of developing colorectal cancer.


Asunto(s)
Competencia Celular , Transformación Celular Neoplásica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Esterasas/metabolismo , Genes APC , Mutación , Adenoma/genética , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Competencia Celular/genética , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Medios de Cultivo Condicionados , Progresión de la Enfermedad , Esterasas/antagonistas & inhibidores , Esterasas/genética , Femenino , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Organoides/citología , Organoides/metabolismo , Organoides/patología , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt
5.
Nat Cell Biol ; 21(11): 1321-1333, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31685987

RESUMEN

Following severe or chronic liver injury, adult ductal cells (cholangiocytes) contribute to regeneration by restoring both hepatocytes and cholangiocytes. We recently showed that ductal cells clonally expand as self-renewing liver organoids that retain their differentiation capacity into both hepatocytes and ductal cells. However, the molecular mechanisms by which adult ductal-committed cells acquire cellular plasticity, initiate organoids and regenerate the damaged tissue remain largely unknown. Here, we describe that ductal cells undergo a transient, genome-wide, remodelling of their transcriptome and epigenome during organoid initiation and in vivo following tissue damage. TET1-mediated hydroxymethylation licences differentiated ductal cells to initiate organoids and activate the regenerative programme through the transcriptional regulation of stem-cell genes and regenerative pathways including the YAP-Hippo signalling. Our results argue in favour of the remodelling of genomic methylome/hydroxymethylome landscapes as a general mechanism by which differentiated cells exit a committed state in response to tissue damage.


Asunto(s)
Proteínas de Unión al ADN/genética , Epigénesis Genética , Epigenoma , Regeneración Hepática/genética , Hígado/metabolismo , Organoides/metabolismo , Proteínas Proto-Oncogénicas/genética , Transcriptoma , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Conductos Biliares/citología , Conductos Biliares/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Vía de Señalización Hippo , Hígado/citología , Masculino , Ratones Transgénicos , Organoides/citología , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteínas Señalizadoras YAP
8.
Nat Commun ; 9(1): 1075, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540680

RESUMEN

A central question in stem cell biology is the relationship between stem cells and their niche. Although previous reports have uncovered how signaling molecules released by niche cells support stem cell function, the role of the extra-cellular matrix (ECM) within the niche is unclear. Here, we show that upon activation, skeletal muscle stem cells (satellite cells) induce local remodeling of the ECM and the deposition of laminin-α1 and laminin-α5 into the basal lamina of the satellite cell niche. Genetic ablation of laminin-α1, disruption of integrin-α6 signaling or blocking matrix metalloproteinase activity impairs satellite cell expansion and self-renewal. Collectively, our findings establish that remodeling of the ECM is an integral process of stem cell activity to support propagation and self-renewal, and may explain the effect laminin-α1-containing supports have on embryonic and adult stem cells, as well as the regenerative activity of exogenous laminin-111 therapy.


Asunto(s)
Autorrenovación de las Células/fisiología , Células Satélite del Músculo Esquelético/citología , Nicho de Células Madre/fisiología , Animales , Membrana Basal/citología , Membrana Basal/metabolismo , Diferenciación Celular/fisiología , Matriz Extracelular/metabolismo , Humanos , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal
9.
Nat Commun ; 9(1): 1020, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29523787

RESUMEN

Cellular senescence is a mechanism that provides an irreversible barrier to cell cycle progression to prevent undesired proliferation. However, under pathological circumstances, senescence can adversely affect organ function, viability and regeneration. We have developed a mouse model of biliary senescence, based on the conditional deletion of Mdm2 in bile ducts under the control of the Krt19 promoter, that exhibits features of biliary disease. Here we report that senescent cholangiocytes induce profound alterations in the cellular and signalling microenvironment, with recruitment of myofibroblasts and macrophages causing collagen deposition, TGFß production and induction of senescence in surrounding cholangiocytes and hepatocytes. Finally, we study how inhibition of TGFß-signalling disrupts the transmission of senescence and restores liver function. We identify cellular senescence as a detrimental mechanism in the development of biliary injury. Our results identify TGFß as a potential therapeutic target to limit senescence-dependent aggravation in human cholangiopathies.


Asunto(s)
Conductos Biliares/lesiones , Conductos Biliares/patología , Senescencia Celular/fisiología , Colangitis Esclerosante/patología , Cirrosis Hepática Biliar/patología , Hígado/patología , Regeneración/fisiología , Animales , Células Cultivadas , Colangitis Esclerosante/terapia , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Hepatocitos/patología , Humanos , Queratina-19/genética , Cirrosis Hepática Biliar/terapia , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo
10.
Nature ; 547(7663): 350-354, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28700576

RESUMEN

After liver injury, regeneration occurs through self-replication of hepatocytes. In severe liver injury, hepatocyte proliferation is impaired-a feature of human chronic liver disease. It is unclear whether other liver cell types can regenerate hepatocytes. Here we use two independent systems to impair hepatocyte proliferation during liver injury to evaluate the contribution of non-hepatocytes to parenchymal regeneration. First, loss of ß1-integrin in hepatocytes with liver injury triggered a ductular reaction of cholangiocyte origin, with approximately 25% of hepatocytes being derived from a non-hepatocyte origin. Second, cholangiocytes were lineage traced with concurrent inhibition of hepatocyte proliferation by ß1-integrin knockdown or p21 overexpression, resulting in the significant emergence of cholangiocyte-derived hepatocytes. We describe a model of combined liver injury and inhibition of hepatocyte proliferation that causes physiologically significant levels of regeneration of functional hepatocytes from biliary cells.


Asunto(s)
Conductos Biliares Intrahepáticos/citología , Hepatocitos/patología , Regeneración Hepática , Hígado/citología , Hígado/patología , Células Madre/citología , Animales , Linaje de la Célula , Proliferación Celular , Femenino , Integrina beta1/genética , Hígado/lesiones , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Proc Natl Acad Sci U S A ; 113(43): 12250-12255, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27791012

RESUMEN

The prognosis of cholangiocarcinoma (CC) is dismal. Notch has been identified as a potential driver; forced exogenous overexpression of Notch1 in hepatocytes results in the formation of biliary tumors. In human disease, however, it is unknown which components of the endogenously signaling pathway are required for tumorigenesis, how these orchestrate cancer, and how they can be targeted for therapy. Here we characterize Notch in human-resected CC, a toxin-driven model in rats, and a transgenic mouse model in which p53 deletion is targeted to biliary epithelia and CC induced using the hepatocarcinogen thioacetamide. We find that across species, the atypical receptor NOTCH3 is differentially overexpressed; it is progressively up-regulated with disease development and promotes tumor cell survival via activation of PI3k-Akt. We use genetic KO studies to show that tumor growth significantly attenuates after Notch3 deletion and demonstrate signaling occurs via a noncanonical pathway independent of the mediator of classical Notch, Recombinant Signal Binding Protein for Immunoglobulin Kappa J Region (RBPJ). These data present an opportunity in this aggressive cancer to selectively target Notch, bypassing toxicities known to be RBPJ dependent.


Asunto(s)
Carcinogénesis/genética , Colangiocarcinoma/genética , Neoplasias Experimentales/genética , Pronóstico , Receptor Notch3/genética , Animales , Colangiocarcinoma/patología , Humanos , Región de Unión de la Inmunoglobulina/genética , Ratones , Ratones Transgénicos , Neoplasias Experimentales/patología , Fosfatidilinositol 3-Quinasas/genética , Ratas , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...