Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34039707

RESUMEN

Specified intestinal epithelial cells reprogram and contribute to the regeneration and renewal of the epithelium upon injury. Mutations that deregulate such renewal processes may contribute to tumorigenesis. Using intestinal organoids, we show that concomitant activation of Notch signaling and ablation of p53 induce a highly proliferative and regenerative cell state, which is associated with increased levels of Yap and the histone methyltransferase Mll1. The induced signaling system orchestrates high proliferation, self-renewal, and niche-factor-independent growth, and elevates the trimethylation of histone 3 at lysine 4 (H3K4me3). We demonstrate that Yap and Mll1 are also elevated in patient-derived colorectal cancer (CRC) organoids and control growth and viability. Our data suggest that Notch activation and p53 ablation induce a signaling circuitry involving Yap and the epigenetic regulator Mll1, which locks cells in a proliferative and regenerative state that renders them susceptible for tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Mutación , Organoides/metabolismo , Factores de Transcripción/metabolismo
2.
Nature ; 523(7558): 92-5, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25970250

RESUMEN

The tumour microenvironment may contribute to tumorigenesis owing to mechanical forces such as fibrotic stiffness or mechanical pressure caused by the expansion of hyper-proliferative cells. Here we explore the contribution of the mechanical pressure exerted by tumour growth onto non-tumorous adjacent epithelium. In the early stage of mouse colon tumour development in the Notch(+)Apc(+/1638N) mouse model, we observed mechanistic pressure stress in the non-tumorous epithelial cells caused by hyper-proliferative adjacent crypts overexpressing active Notch, which is associated with increased Ret and ß-catenin signalling. We thus developed a method that allows the delivery of a defined mechanical pressure in vivo, by subcutaneously inserting a magnet close to the mouse colon. The implanted magnet generated a magnetic force on ultra-magnetic liposomes, stabilized in the mesenchymal cells of the connective tissue surrounding colonic crypts after intravenous injection. The magnetically induced pressure quantitatively mimicked the endogenous early tumour growth stress in the order of 1,200 Pa, without affecting tissue stiffness, as monitored by ultrasound strain imaging and shear wave elastography. The exertion of pressure mimicking that of tumour growth led to rapid Ret activation and downstream phosphorylation of ß-catenin on Tyr654, imparing its interaction with the E-cadherin in adherens junctions, and which was followed by ß-catenin nuclear translocation after 15 days. As a consequence, increased expression of ß-catenin-target genes was observed at 1 month, together with crypt enlargement accompanying the formation of early tumorous aberrant crypt foci. Mechanical activation of the tumorigenic ß-catenin pathway suggests unexplored modes of tumour propagation based on mechanical signalling pathways in healthy epithelial cells surrounding the tumour, which may contribute to tumour heterogeneity.


Asunto(s)
Carcinogénesis/patología , Neoplasias del Colon/fisiopatología , Presión , Microambiente Tumoral , beta Catenina/genética , Transporte Activo de Núcleo Celular , Animales , Células Epiteliales/citología , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Imanes , Masculino , Nanopartículas del Metal , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , beta Catenina/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(2): E283-E291, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29279402

RESUMEN

Colonization by Streptococcus gallolyticus subsp. gallolyticus (SGG) is strongly associated with the occurrence of colorectal cancer (CRC). However, the factors leading to its successful colonization are unknown, and whether SGG influences the oncogenic process or benefits from the tumor-prone environment to prevail remains an open question. Here, we elucidate crucial steps that explain how CRC favors SGG colonization. By using mice genetically prone to CRC, we show that SGG colonization is 1,000-fold higher in tumor-bearing mice than in normal mice. This selective advantage occurs at the expense of resident intestinal enterococci. An SGG-specific locus encoding a bacteriocin ("gallocin") is shown to kill enterococci in vitro. Importantly, bile acids strongly enhance this bacteriocin activity in vivo, leading to greater SGG colonization. Constitutive activation of the Wnt pathway, one of the earliest signaling alterations in CRC, and the decreased expression of the bile acid apical transporter gene Slc10A2, as an effect of the Apc founding mutation, may thereby sustain intestinal colonization by SGG. We conclude that CRC-specific conditions promote SGG colonization of the gut by replacing commensal enterococci in their niche.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Tracto Gastrointestinal/microbiología , Streptococcus gallolyticus/fisiología , Adenoma , Animales , Bacteriocinas/genética , Bacteriocinas/metabolismo , Ácidos y Sales Biliares/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Simportadores/genética , Simportadores/metabolismo
4.
Biol Cell ; 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29907957

RESUMEN

BACKGROUND INFORMATION: Tumor stroma remodeling is a key feature of malignant tumors and can promote cancer progression. Laminins are major constituents of basement membranes that physically separate the epithelium from the underlying stroma. RESULTS: By employing mouse models expressing high and low levels of the laminin α1 chain (LMα1), we highlighted its implication in a tumor-stroma crosstalk, thus leading to increased colon tumor incidence, angiogenesis and tumor growth. The underlying mechanism involves attraction of carcinoma-associated fibroblasts by LMα1, VEGFA expression triggered by the complex integrin α2ß1-CXCR4 and binding of VEGFA to LM-111, which in turn promotes angiogenesis, tumor cell survival and proliferation. A gene signature comprising LAMA1, ITGB1, ITGA2, CXCR4 and VEGFA has negative predictive value in colon cancer. CONCLUSIONS: Together, we have identified VEGFA, CXCR4 and α2ß1 integrin downstream of LMα1 in colon cancer as of bad prognostic value for patient survival. SIGNIFICANCE: This information opens novel opportunities for diagnosis and treatment of colon cancer.

5.
PLoS Genet ; 12(10): e1006349, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27736879

RESUMEN

Zinc transporters play a critical role in spatiotemporal regulation of zinc homeostasis. Although disruption of zinc homeostasis has been implicated in disorders such as intestinal inflammation and aberrant epithelial morphology, it is largely unknown which zinc transporters are responsible for the intestinal epithelial homeostasis. Here, we show that Zrt-Irt-like protein (ZIP) transporter ZIP7, which is highly expressed in the intestinal crypt, is essential for intestinal epithelial proliferation. Mice lacking Zip7 in intestinal epithelium triggered endoplasmic reticulum (ER) stress in proliferative progenitor cells, leading to significant cell death of progenitor cells. Zip7 deficiency led to the loss of Olfm4+ intestinal stem cells and the degeneration of post-mitotic Paneth cells, indicating a fundamental requirement for Zip7 in homeostatic intestinal regeneration. Taken together, these findings provide evidence for the importance of ZIP7 in maintenance of intestinal epithelial homeostasis through the regulation of ER function in proliferative progenitor cells and maintenance of intestinal stem cells. Therapeutic targeting of ZIP7 could lead to effective treatment of gastrointestinal disorders.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proliferación Celular/genética , Estrés del Retículo Endoplásmico/genética , Zinc/metabolismo , Animales , Apoptosis/genética , Proteínas de Transporte de Catión/biosíntesis , Retículo Endoplásmico/genética , Células Epiteliales/metabolismo , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/metabolismo , Técnicas de Inactivación de Genes , Homeostasis , Mucosa Intestinal/crecimiento & desarrollo , Mucosa Intestinal/metabolismo , Ratones , Organoides/crecimiento & desarrollo , Células de Paneth/metabolismo , Células Madre/metabolismo
6.
Gut ; 66(10): 1748-1760, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27371534

RESUMEN

OBJECTIVE: Epidemiological and clinical data indicate that patients suffering from IBD with long-standing colitis display a higher risk to develop colorectal high-grade dysplasia. Whereas carcinoma invasion and metastasis rely on basement membrane (BM) disruption, experimental evidence is lacking regarding the potential contribution of epithelial cell/BM anchorage on inflammation onset and subsequent neoplastic transformation of inflammatory lesions. Herein, we analyse the role of the α6ß4 integrin receptor found in hemidesmosomes that attach intestinal epithelial cells (IECs) to the laminin-containing BM. DESIGN: We developed new mouse models inducing IEC-specific ablation of α6 integrin either during development (α6ΔIEC) or in adults (α6ΔIEC-TAM). RESULTS: Strikingly, all α6ΔIEC mutant mice spontaneously developed long-standing colitis, which degenerated overtime into infiltrating adenocarcinoma. The sequence of events leading to disease onset entails hemidesmosome disruption, BM detachment, IL-18 overproduction by IECs, hyperplasia and enhanced intestinal permeability. Likewise, IEC-specific ablation of α6 integrin induced in adult mice (α6ΔIEC-TAM) resulted in fully penetrant colitis and tumour progression. Whereas broad-spectrum antibiotic treatment lowered tissue pathology and IL-1ß secretion from infiltrating myeloid cells, it failed to reduce Th1 and Th17 response. Interestingly, while the initial intestinal inflammation occurred independently of the adaptive immune system, tumourigenesis required B and T lymphocyte activation. CONCLUSIONS: We provide for the first time evidence that loss of IECs/BM interactions triggered by hemidesmosome disruption initiates the development of inflammatory lesions that progress into high-grade dysplasia and carcinoma. Colorectal neoplasia in our mouse models resemble that seen in patients with IBD, making them highly attractive for discovering more efficient therapies.


Asunto(s)
Adenocarcinoma/fisiopatología , Colitis/fisiopatología , Neoplasias Colorrectales/fisiopatología , Citocinas/metabolismo , Hemidesmosomas/fisiología , Integrina alfa6/genética , Integrina alfa6beta4/metabolismo , Mucosa Intestinal/metabolismo , Inmunidad Adaptativa , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Linfocitos B , Membrana Basal/fisiopatología , Caspasa 1/metabolismo , Colitis/genética , Colitis/metabolismo , Colitis/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Citocinas/genética , Células Epiteliales/metabolismo , Hemidesmosomas/genética , Homeostasis/genética , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Queratina-18/metabolismo , Queratina-8/metabolismo , Activación de Linfocitos , Ratones , Moco/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Permeabilidad , Índice de Severidad de la Enfermedad , Transducción de Señal , Linfocitos T
7.
Proc Natl Acad Sci U S A ; 111(21): E2229-36, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24825889

RESUMEN

In the majority of microsatellite-stable colorectal cancers (CRCs), an initiating mutation occurs in the adenomatous polyposis coli (APC) or ß-catenin gene, activating the ß-catenin/TCF pathway. The progression of resulting adenomas is associated with oncogenic activation of KRas and inactivation of the p53 and TGF-ß/Smad functions. Most established CRC cell lines contain mutations in the TGF-ß/Smad pathway, but little is known about the function of TGF-ß in the early phases of intestinal tumorigenesis. We used mouse and human ex vivo 3D intestinal organoid cultures and in vivo mouse models to study the effect of TGF-ß on the Lgr5(+) intestinal stem cells and their progeny in intestinal adenomas. We found that the TGF-ß-induced apoptosis in Apc-mutant organoids, including the Lgr5(+) stem cells, was mediated by up-regulation of the BH3-only proapoptotic protein Bcl-2-like protein 11 (Bim). BH3-mimetic compounds recapitulated the effect of Bim not only in the adenomas but also in human CRC organoids that had lost responsiveness to TGF-ß-induced apoptosis. However, wild-type intestinal crypts were markedly less sensitive to TGF-ß than Apc-mutant adenomas, whereas the KRas oncogene increased resistance to TGF-ß via the activation of the Erk1/2 kinase pathway, leading to Bim down-regulation. Our studies identify Bim as a critical mediator of TGF-ß-induced apoptosis in intestinal adenomas and show that the common progression mutations modify Bim levels and sensitivity to TGF-ß during intestinal adenoma development.


Asunto(s)
Adenoma/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Intestinales/genética , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteína 11 Similar a Bcl2 , Western Blotting , Células Cultivadas , Cromatografía en Gel , Cartilla de ADN/genética , Citometría de Flujo , Humanos , Ratones , Análisis por Micromatrices , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
8.
PLoS Comput Biol ; 11(11): e1004571, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26528548

RESUMEN

Understanding the etiology of metastasis is very important in clinical perspective, since it is estimated that metastasis accounts for 90% of cancer patient mortality. Metastasis results from a sequence of multiple steps including invasion and migration. The early stages of metastasis are tightly controlled in normal cells and can be drastically affected by malignant mutations; therefore, they might constitute the principal determinants of the overall metastatic rate even if the later stages take long to occur. To elucidate the role of individual mutations or their combinations affecting the metastatic development, a logical model has been constructed that recapitulates published experimental results of known gene perturbations on local invasion and migration processes, and predict the effect of not yet experimentally assessed mutations. The model has been validated using experimental data on transcriptome dynamics following TGF-ß-dependent induction of Epithelial to Mesenchymal Transition in lung cancer cell lines. A method to associate gene expression profiles with different stable state solutions of the logical model has been developed for that purpose. In addition, we have systematically predicted alleviating (masking) and synergistic pairwise genetic interactions between the genes composing the model with respect to the probability of acquiring the metastatic phenotype. We focused on several unexpected synergistic genetic interactions leading to theoretically very high metastasis probability. Among them, the synergistic combination of Notch overexpression and p53 deletion shows one of the strongest effects, which is in agreement with a recent published experiment in a mouse model of gut cancer. The mathematical model can recapitulate experimental mutations in both cell line and mouse models. Furthermore, the model predicts new gene perturbations that affect the early steps of metastasis underlying potential intervention points for innovative therapeutic strategies in oncology.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Modelos Biológicos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Animales , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Biología Computacional , Bases de Datos Genéticas , Humanos , Ratones , Ratones Transgénicos , Mutación
9.
Proc Natl Acad Sci U S A ; 110(15): E1380-9, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23520048

RESUMEN

Efficient wound healing is required to maintain the integrity of the intestinal epithelial barrier because of its constant exposure to a large variety of environmental stresses. This process implies a partial cell depolarization and the acquisition of a motile phenotype that involves rearrangements of the actin cytoskeleton. Here we address how polarized enterocytes harboring actin-rich apical microvilli undergo extensive cell remodeling to drive injury repair. Using live imaging technologies, we demonstrate that enterocytes in vitro and in vivo rapidly depolarize their microvilli at the wound edge. Through its F-actin-severing activity, the microvillar actin-binding protein villin drives both apical microvilli disassembly in vitro and in vivo and promotes lamellipodial extension. Photoactivation experiments indicate that microvillar actin is mobilized at the lamellipodium, allowing optimal migration. Finally, efficient repair of colonic mechanical injuries requires villin severing of F-actin, emphasizing the importance of villin function in intestinal homeostasis. Thus, villin severs F-actin to ensure microvillus depolarization and enterocyte remodeling upon injury. This work highlights the importance of specialized apical pole disassembly for the repolarization of epithelial cells initiating migration.


Asunto(s)
Actinas/química , Enterocitos/citología , Proteínas de Microfilamentos/fisiología , Actinas/metabolismo , Animales , Apoptosis , Diferenciación Celular , Línea Celular , Movimiento Celular , Proliferación Celular , Endoscopía , Enterocitos/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microvellosidades/metabolismo , Fenotipo , Porcinos , Cicatrización de Heridas
10.
Dev Biol ; 387(2): 191-202, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24440658

RESUMEN

Krüppel-like factor 5 (KLF5) is a pro-proliferative transcriptional regulator primarily expressed in the intestinal crypt epithelial cells. Constitutive intestine-specific deletion of Klf5 is neonatal lethal suggesting a crucial role for KLF5 in intestinal development and homeostasis. We have previously shown Klf5 to play an active role regulating intestinal tumorigenesis. Here we examine the effect of inducible intestine-specific deletion of Klf5 in adult mice. Klf5 is lost from the intestine beginning at day 3 after the start of a 5-day treatment with the inducer tamoxifen. Although the mice have no significant weight loss or lethality, the colonic tissue shows signs of epithelial distress starting at day 3 following induction. Accompanying the morphological changes is a significant loss of proliferative crypt epithelial cells as revealed by BrdU or Ki67 staining at days 3 and 5 after start of tamoxifen. We also observed a loss of goblet cells from the colon and Paneth cells from the small intestine upon induced deletion of Klf5. In addition, loss of Klf5 from the colonic epithelium is accompanied by a regenerative response that coincides with an expansion in the zone of Sox9 expression along the crypt axis. At day 11, both proliferation and Sox9 expression return to baseline levels. Microarray and quantitative PCR analyses reveal an up-regulation of several regeneration-associated genes (Reg1A, Reg3G and Reg3B) and down-regulation of many Klf5 targets (Ki-67, cyclin B, Cdc2 and cyclin D1). Sox9 and Reg1A protein levels are also increased upon Klf5 loss. Lentiviral-mediated knockdown of KLF5 and exogenous expression of KLF5 in colorectal cancer cell lines confirm that Sox9 expression is negatively regulated by KLF5. Furthermore, ChIP assays reveal a direct association of KLF5 with both the Sox9 and Reg1A promoters. We have shown that disruption of epithelial homeostasis due to Klf5 loss from the adult colon is followed by a regenerative response led by Sox9 and the Reg family of proteins. Our study demonstrates that adult mouse colonic tissue undergoes acute physiological changes to accommodate the loss of Klf5 withstanding epithelial damage further signifying importance of Klf5 in colonic homeostasis.


Asunto(s)
Colon/fisiología , Factores de Transcripción de Tipo Kruppel/genética , Regeneración/genética , Animales , Antineoplásicos Hormonales/farmacología , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Ciclina B/metabolismo , Ciclina D1/metabolismo , Regulación hacia Abajo , Células Caliciformes/efectos de los fármacos , Células HCT116 , Células HEK293 , Humanos , Antígeno Ki-67/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Litostatina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis , Células de Paneth/efectos de los fármacos , Regiones Promotoras Genéticas , Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción SOX9/metabolismo , Eliminación de Secuencia , Transducción de Señal/genética , Tamoxifeno/farmacología , Regulación hacia Arriba
11.
Ann Surg Oncol ; 22 Suppl 3: S1475-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25956578

RESUMEN

BACKGROUND: Colonic self-expanding metallic stents (SEMS) are used in obstructive colorectal cancer patients as a bridge to surgery. However, its oncologic safety remains uncertain. Therefore, we attempted to clarify this further with an experimental study and constructed a mouse model of colonic cancer. METHODS: CT26 cells were injected in the rectal wall, and to mimic SEMS, a cardiac stent was inserted under endoscopy in occlusive (75 % lumen occlusion) tumors. We set up a control group (n = 22) and a stent group (n = 16), and the findings were compared. We focused on serum lactate dehydrogenase (LDH) concentrations, circulating tumor cells, survival time, peritoneal carcinomatosis, liver metastases, and bioluminescence. RESULTS: One week after stent insertion, the serum LDH concentrations were significantly higher in the stent group (506 ± 203 IU/L) compared to the controls (229 ± 52 IU/L) (P = 0.005). The average survival time before sacrifice was significantly lower in the stent group (15.2 ± 1 days) compared to the controls (20 ± 5 days) (P = 0.005). The presence of a peritoneal carcinomatosis was more frequently observed in the stent group (75 %) than in the controls (50 %). Liver metastases were observed in 19 % of the stent group compared to the controls (4.5 %) (P = 0.29). After multivariate analysis, the stent group was still found to be associated with significantly lower survival time (P = 0.002). CONCLUSIONS: These observations led us to conclude that in our mouse model, SEMS resulted in an increased metastatic process and a shorter survival time. We suggest, therefore, that the utmost caution be exercised when opting for a stent as a bridge to surgery.


Asunto(s)
Neoplasias del Colon/cirugía , Modelos Animales de Enfermedad , Obstrucción Intestinal/cirugía , Neoplasias Hepáticas/secundario , Neoplasias Peritoneales/secundario , Stents/efectos adversos , Animales , Neoplasias del Colon/patología , Humanos , Ratones
12.
J Biol Chem ; 287(39): 32598-616, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22851168

RESUMEN

Glycosphingolipids (GSLs) constitute major components of enterocytes and were hypothesized to be potentially important for intestinal epithelial polarization. The enzyme UDP-glucose ceramide glucosyltransferase (Ugcg) catalyzes the initial step of GSL biosynthesis. Newborn and adult mice with enterocyte-specific genetic deletion of the gene Ugcg were generated. In newborn mutants lacking GSLs at day P0, intestinal epithelia were indistinguishable from those in control littermates displaying an intact polarization with regular brush border. However, those mice were not consistently able to absorb nutritional lipids from milk. Between postnatal days 5 and 7, severe defects in intestinal epithelial differentiation occurred accompanied by impaired intestinal uptake of nutrients. Villi of mutant mice became stunted, and enterocytes lacked brush border. The defects observed in mutant mice caused diarrhea, malabsorption, and early death. In this study, we show that GSLs are essential for enterocyte resorptive function but are primarily not for polarization; GSLs are required for intracellular vesicular transport in resorption-active intestine.


Asunto(s)
Polaridad Celular/fisiología , Enterocitos/metabolismo , Glucosiltransferasas/metabolismo , Glicoesfingolípidos/biosíntesis , Absorción Intestinal/fisiología , Animales , Equidae , Eliminación de Gen , Glucosiltransferasas/genética , Glicoesfingolípidos/genética , Cabras , Ratones , Ratones Mutantes , Conejos
13.
Development ; 137(9): 1573-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20388655

RESUMEN

The intestinal epithelium is a complex system characterized by massive and continuous cell renewal and differentiation. In this context, cell-type-specific transcription factors are thought to play a crucial role by modulating specific transcription networks and signalling pathways. Hnf1alpha and beta are closely related atypical homeoprotein transcription factors expressed in several epithelia, including the gut. With the use of a conditional inactivation system, we generated mice in which Hnf1b is specifically inactivated in the intestinal epithelium on a wild-type or Hnf1a(-/-) genetic background. Whereas the inactivation of Hnf1a or Hnf1b alone did not lead to any major intestinal dysfunction, the concomitant inactivation of both genes resulted in a lethal phenotype. Double-mutant animals had defective differentiation and cell fate commitment. The expression levels of markers of all the differentiated cell types, both enterocytes and secretory cells, were affected. In addition, the number of goblet cells was increased, whereas mature Paneth cells were missing. At the molecular level, we show that Hnf1alpha and beta act upstream of the Notch pathway controlling directly the expression of two crucial components: Jag1 and Atoh1. We demonstrate that the double-mutant mice present with a defect in intestinal water absorption and that Hnf1alpha and beta directly control the expression of Slc26a3, a gene whose mutations are associated with chloride diarrhoea in human patients. Our study identifies new direct target genes of the Hnf1 transcription factors and shows that they play crucial roles in both defining cell fate and controlling terminal functions in the gut epithelium.


Asunto(s)
Diferenciación Celular/fisiología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Animales , Antiportadores/genética , Antiportadores/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/genética , Inmunoprecipitación de Cromatina , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/genética , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Proteínas Serrate-Jagged , Transportadores de Sulfato
14.
J Cell Biol ; 178(4): 635-48, 2007 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-17698607

RESUMEN

The HMG-box transcription factor Sox9 is expressed in the intestinal epithelium, specifically, in stem/progenitor cells and in Paneth cells. Sox9 expression requires an active beta-catenin-Tcf complex, the transcriptional effector of the Wnt pathway. This pathway is critical for numerous aspects of the intestinal epithelium physiopathology, but processes that specify the cell response to such multipotential signals still remain to be identified. We inactivated the Sox9 gene in the intestinal epithelium to analyze its physiological function. Sox9 inactivation affected differentiation throughout the intestinal epithelium, with a disappearance of Paneth cells and a decrease of the goblet cell lineage. Additionally, the morphology of the colon epithelium was severely altered. We detected general hyperplasia and local crypt dysplasia in the intestine, and Wnt pathway target genes were up-regulated. These results highlight the central position of Sox9 as both a transcriptional target and a regulator of the Wnt pathway in the regulation of intestinal epithelium homeostasis.


Asunto(s)
Colon/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Células de Paneth/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Colon/citología , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Células de Paneth/citología , Factor de Transcripción SOX9 , Factores de Transcripción/genética
15.
Exp Cell Res ; 317(19): 2740-7, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21745469

RESUMEN

Notch signaling has been recently shown to have a fundamental role in stem cell maintenance and control of proper homeostasis in the intestine of different species. Here, we briefly review the current literature on Notch signals in the intestine of Drosophila, Zebrafish and the mouse, and try to highlight conserved and divergent Notch functions across species. Notch signals show a remarkably conserved role in skewing cell fate choices in intestinal lineages throughout evolution. Genetic analysis demonstrates that loss of Notch signaling invariably leads to increased numbers of secretory cells and loss of enterocytes, while gain of Notch function will completely block secretory cell differentiation. Finally, we discuss the potential contribution of Notch signaling to the initiation of colorectal cancer by controlling the maintenance of the undifferentiated state of intestinal neoplastic cells and speculate on the therapeutic consequences of affecting cancer stem cells.


Asunto(s)
Drosophila , Homeostasis/genética , Intestinos/fisiología , Ratones , Receptores Notch/fisiología , Pez Cebra , Animales , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiología , Homeostasis/fisiología , Humanos , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Ratones/genética , Ratones/metabolismo , Ratones/fisiología , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Especificidad de la Especie , Pez Cebra/genética , Pez Cebra/metabolismo , Pez Cebra/fisiología
16.
Proc Natl Acad Sci U S A ; 106(15): 6309-14, 2009 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-19251639

RESUMEN

Notch and Wnt signals play essential roles in intestinal development and homeostasis, yet how they integrate their action to affect intestinal morphogenesis is not understood. We examined the interplay between these two signaling pathways in vivo, by modulating Notch activity in mice carrying either a loss- or a gain-of-function mutation of Wnt signaling. We find that the dramatic proliferative effect that Notch signals have on early intestinal precursors requires normal Wnt signaling, whereas its influence on intestinal differentiation appears independent of Wnt. Analogous experiments in Drosophila demonstrate that the synergistic effects of Notch and Wnt are valid across species. We also demonstrate a striking synergy between Notch and Wnt signals that results in inducing the formation of intestinal adenomas, particularly in the colon, a region rarely affected in available mouse tumor models, but the primary target organ in human patients. These studies thus reveal a previously unknown oncogenic potential of Notch signaling in colorectal tumorigenesis that, significantly, is supported by the analysis of human tumors. Importantly, our experimental evidence raises the possibility that Notch activation might be an essential initial event triggering colorectal cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Receptores Notch/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Neoplasias Intestinales/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores Notch/genética , Tasa de Supervivencia , Factores de Transcripción TCF/deficiencia , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Factor de Transcripción 4 , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
17.
Nat Commun ; 13(1): 1500, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35314700

RESUMEN

Although the mammalian intestinal epithelium manifests robust regenerative capacity after various cytotoxic injuries, the underlying mechanism has remained unclear. Here we identify the cyclin-dependent kinase inhibitor p57 as a specific marker for a quiescent cell population located around the +4 position of intestinal crypts. Lineage tracing reveals that the p57+ cells serve as enteroendocrine/tuft cell precursors under normal conditions but dedifferentiate and act as facultative stem cells to support regeneration after injury. Single-cell transcriptomics analysis shows that the p57+ cells undergo a dynamic reprogramming process after injury that is characterized by fetal-like conversion and metaplasia-like transformation. Population-level analysis also detects such spatiotemporal reprogramming widely in other differentiated cell types. In intestinal adenoma, p57+ cells manifest homeostatic stem cell activity, in the context of constitutively activated spatiotemporal reprogramming. Our results highlight a pronounced plasticity of the intestinal epithelium that supports maintenance of tissue integrity in normal and neoplastic contexts.


Asunto(s)
Mucosa Intestinal , Neoplasias , Animales , Diferenciación Celular , Mucosa Intestinal/metabolismo , Intestinos , Mamíferos , Neoplasias/metabolismo , Células Madre/metabolismo
18.
Nature ; 435(7044): 964-8, 2005 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15959516

RESUMEN

The Notch signalling pathway plays a crucial role in specifying cellular fates in metazoan development by regulating communication between adjacent cells. Correlative studies suggested an involvement of Notch in intestinal development. Here, by modulating Notch activity in the mouse intestine, we directly implicate Notch signals in intestinal cell lineage specification. We also show that Notch activation is capable of amplifying the intestinal progenitor pool while inhibiting cell differentiation. We conclude that Notch activity is required for the maintenance of proliferating crypt cells in the intestinal epithelium.


Asunto(s)
Linaje de la Célula , Mucosa Intestinal/metabolismo , Intestinos/citología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Notch1 , Receptores de Superficie Celular/genética , Factor de Transcripción HES-1 , Factores de Transcripción/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
19.
Nature ; 435(7044): 959-63, 2005 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15959515

RESUMEN

The self-renewing epithelium of the small intestine is ordered into stem/progenitor crypt compartments and differentiated villus compartments. Recent evidence indicates that the Wnt cascade is the dominant force in controlling cell fate along the crypt-villus axis. Here we show a rapid, massive conversion of proliferative crypt cells into post-mitotic goblet cells after conditional removal of the common Notch pathway transcription factor CSL/RBP-J. We obtained a similar phenotype by blocking the Notch cascade with a gamma-secretase inhibitor. The inhibitor also induced goblet cell differentiation in adenomas in mice carrying a mutation of the Apc tumour suppressor gene. Thus, maintenance of undifferentiated, proliferative cells in crypts and adenomas requires the concerted activation of the Notch and Wnt cascades. Our data indicate that gamma-secretase inhibitors, developed for Alzheimer's disease, might be of therapeutic benefit in colorectal neoplastic disease.


Asunto(s)
Adenoma/patología , Proliferación Celular/efectos de los fármacos , Endopeptidasas/metabolismo , Células Caliciformes/citología , Intestino Delgado/citología , Proteínas de la Membrana/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Adenoma/enzimología , Adenoma/genética , Adenoma/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico Endopeptidasas , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dibenzazepinas/farmacología , Femenino , Genes APC , Células Caliciformes/efectos de los fármacos , Células Caliciformes/metabolismo , Células Caliciformes/patología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Intestino Delgado/efectos de los fármacos , Intestino Delgado/enzimología , Intestino Delgado/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Receptores Notch , Transducción de Señal/efectos de los fármacos
20.
Proc Natl Acad Sci U S A ; 105(39): 15058-63, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18815378

RESUMEN

NF-kappaB is a key transcriptional regulator of inflammatory responses, but also controls expression of prosurvival genes, whose products protect tissues from damage and may thus act indirectly in an antiinflammatory fashion. The variable importance of these two distinct NF-kappaB-controlled responses impacts the potential utility of NF-kappaB inhibition as a treatment strategy for intractable inflammatory conditions, such as inflammatory bowel disease. Here, we show in murine models that inhibition of IKKbeta-dependent NF-kappaB activation exacerbates acute inflammation, but attenuates chronic inflammatory disease in the intestinal tract. Acute ulcerating inflammation is aggravated because of diminished NF-kappaB-mediated protection against epithelial cell apoptosis and delayed mucosal regeneration secondary to reduced NF-kappaB-dependent recruitment of inflammatory cells that secrete cytoprotective factors. In contrast, in IL-10-deficient mice, which serve as a model of chronic T cell-dependent colitis, ablation of IKKbeta in the intestinal epithelium has no impact, yet IKKbeta deficiency in myeloid cells attenuates inflammation and prolongs survival. These results highlight the striking context and tissue dependence of the proinflammatory and antiapoptotic functions of NF-kappaB. Our findings caution against the therapeutic use of IKKbeta/NF-kappaB inhibitors in acute inflammatory settings dominated by cell loss and ulceration.


Asunto(s)
Colitis Ulcerosa/metabolismo , Quinasa I-kappa B/metabolismo , Enfermedad Aguda , Animales , Enfermedad Crónica , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Expresión Génica , Quinasa I-kappa B/antagonistas & inhibidores , Interleucina-1/genética , Mucosa Intestinal , Ratones , Ratones Mutantes , FN-kappa B/metabolismo , Factor de Transcripción STAT3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA