Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(5): 1704-1713, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30635419

RESUMEN

Inactivating mutations of Arid1a, a subunit of the Switch/sucrose nonfermentable chromatin remodeling complex, have been reported in multiple human cancers. Intestinal deletion of Arid1a has been reported to induce colorectal cancer in mice; however, its functional role in intestinal homeostasis remains unclear. We investigated the functional role of Arid1a in intestinal homeostasis in mice. We found that intestinal deletion of Arid1a results in loss of intestinal stem cells (ISCs), decreased Paneth and goblet cells, disorganized crypt-villous structures, and increased apoptosis in adult mice. Spheroids did not develop from intestinal epithelial cells deficient for Arid1a Lineage-tracing experiments revealed that Arid1a deletion in Lgr5+ ISCs leads to impaired self-renewal of Lgr5+ ISCs but does not perturb intestinal homeostasis. The Wnt signaling pathway, including Wnt agonists, receptors, and target genes, was strikingly down-regulated in Arid1a-deficient intestines. We found that Arid1a directly binds to the Sox9 promoter to support its expression. Remarkably, overexpression of Sox9 in intestinal epithelial cells abrogated the above phenotypes, although Sox9 overexpression in intestinal epithelial cells did not restore the expression levels of Wnt agonist and receptor genes. Furthermore, Sox9 overexpression permitted development of spheroids from Arid1a-deficient intestinal epithelial cells. In addition, deletion of Arid1a concomitant with Sox9 overexpression in Lgr5+ ISCs restores self-renewal in Arid1a-deleted Lgr5+ ISCs. These results indicate that Arid1a is indispensable for the maintenance of ISCs and intestinal homeostasis in mice. Mechanistically, this is mainly mediated by Sox9. Our data provide insights into the molecular mechanisms underlying maintenance of ISCs and intestinal homeostasis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Nucleares/metabolismo , Factor de Transcripción SOX9/metabolismo , Células Madre/metabolismo , Animales , Células Epiteliales/metabolismo , Homeostasis/fisiología , Intestinos/fisiología , Ratones , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción , Vía de Señalización Wnt/fisiología
2.
J Pathol ; 248(2): 179-190, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30689202

RESUMEN

In the mammalian stomach, the isthmus has been considered as a stem cell zone. However, various locations and proliferative activities of gastric stem cells have been reported. We focused here on the stem cell marker Bmi1, a polycomb group protein, aiming to elucidate the characteristics of Bmi1-expressing cells in the stomach and to examine their stem cell potential. We investigated the Bmi1-expressing cell lineage in Bmi1-CreERT; Rosa26-YFP, LacZ or Rosa26-Confetti mice. We examined the in vivo and ex vivo effects of Bmi1-expressing cell ablation by using Bmi1-CreERT; Rosa26-iDTR mice. The Bmi1 lineage was also traced during regeneration after high-dose tamoxifen-, irradiation- and acetic acid-induced mucosal injuries. In the lineage-tracing experiments using low-dose tamoxifen, Bmi1-expressing cells in the isthmus of the gastric antrum and corpus provided progeny bidirectionally, towards both the luminal and basal sides over 6 months. In gastric organoids, Bmi1-expressing cells also provided progeny. Ablation of Bmi1-expressing cells resulted in impaired gastric epithelium in both mouse stomach and organoids. After high-dose tamoxifen-induced gastric mucosal injury, Bmi1-expressing cell lineages expanded and fully occupied all gastric glands of the antrum and the corpus within 7 days after tamoxifen injection. After irradiation- and acetic acid-induced gastric mucosal injuries, Bmi1-expressing cells also contributed to regeneration. In conclusion, Bmi1 is a gastric stem cell marker expressed in the isthmus of the antrum and corpus. Bmi1-expressing cells have stem cell potentials, both under physiological conditions and during regeneration after gastric mucosal injuries. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Antro Pilórico/metabolismo , Células Madre/metabolismo , Ácido Acético , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Modelos Animales de Enfermedad , Fluorouracilo/toxicidad , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones Transgénicos , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Antro Pilórico/efectos de los fármacos , Antro Pilórico/embriología , Antro Pilórico/efectos de la radiación , Regeneración , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/efectos de la radiación , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/metabolismo , Úlcera Gástrica/patología , Tamoxifeno/toxicidad
3.
Gastroenterology ; 155(1): 194-209.e2, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29604291

RESUMEN

BACKGROUND & AIMS: The ARID1A gene encodes a protein that is part of the large adenosine triphosphate (ATP)-dependent chromatin remodeling complex SWI/SNF and is frequently mutated in human pancreatic ductal adenocarcinomas (PDACs). We investigated the functions of ARID1A during formation of PDACs in mice. METHODS: We performed studies with Ptf1a-Cre;KrasG12D mice, which express activated Kras in the pancreas and develop pancreatic intraepithelial neoplasias (PanINs), as well as those with disruption of Aird1a (Ptf1a-Cre;KrasG12D;Arid1af/f mice) or disruption of Brg1 (encodes a catalytic ATPase of the SWI/SNF complex) (Ptf1a-Cre;KrasG12D; Brg1f/fmice). Pancreatic ductal cells (PDCs) were isolated from Arid1af/f mice and from Arid1af/f;SOX9OE mice, which overexpress human SOX9 upon infection with an adenovirus-expressing Cre recombinase. Pancreatic tissues were collected from all mice and analyzed by histology and immunohistochemistry; cells were isolated and grown in 2-dimensional and 3-dimensional cultures. We performed microarray analyses to compare gene expression patterns in intraductal papillary mucinous neoplasms (IPMNs) from the different strains of mice. We obtained 58 samples of IPMNs and 44 samples of PDACs from patients who underwent pancreatectomy in Japan and analyzed them by immunohistochemistry. RESULTS: Ptf1a-Cre;KrasG12D mice developed PanINs, whereas Ptf1a-Cre;KrasG12D;Arid1af/f mice developed IPMNs and PDACs; IPMNs originated from PDCs. ARID1A-deficient IPMNs did not express SOX9. ARID1A-deficient PDCs had reduced expression of SOX9 and dedifferentiated in culture. Overexpression of SOX9 in these cells allowed them to differentiate and prevented dilation of ducts. Among mice with pancreatic expression of activated Kras, those with disruption of Arid1a developed fewer PDACs from IPMNs than mice with disruption of Brg1. ARID1A-deficient IPMNs had reduced activity of the mTOR pathway. Human IPMN and PDAC specimens had reduced levels of ARID1A, SOX9, and phosphorylated S6 (a marker of mTOR pathway activation). Levels of ARID1A correlated with levels of SOX9 and phosphorylated S6. CONCLUSIONS: ARID1A regulates expression of SOX9, activation of the mTOR pathway, and differentiation of PDCs. ARID1A inhibits formation of PDACs from IPMNs in mice with pancreatic expression of activated KRAS and is down-regulated in IPMN and PDAC tissues from patients.


Asunto(s)
Adenocarcinoma in Situ/genética , Carcinoma Ductal Pancreático/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Conductos Pancreáticos/citología , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor de Transcripción SOX9/genética , Adenocarcinoma in Situ/metabolismo , Animales , Carcinogénesis/genética , Carcinoma Ductal Pancreático/metabolismo , Técnicas de Cultivo de Célula , Ratones , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción
5.
Nihon Shokakibyo Gakkai Zasshi ; 111(7): 1416-23, 2014 Jul.
Artículo en Japonés | MEDLINE | ID: mdl-24998733

RESUMEN

A 64-year-old man presented with diarrhea, fever, and disturbance of consciousness; he was subsequently diagnosed with acute renal and hepatic disorder. Abdominal computed tomography identified a gas-forming liver abscess, and the patient underwent emergency drainage. However, his condition did not improve, and Clostridium perfringens was observed in his blood culture. Continuous perfusion drainage was performed by placing an additional drainage tube, which resulted in abscess shrinkage and improved the patient's general condition. Despite the low survival rate in patients with gas-forming liver abscesses caused by C. perfringens, therapy was successful in this patient.


Asunto(s)
Infecciones por Clostridium/complicaciones , Clostridium perfringens , Drenaje/métodos , Absceso Hepático/etiología , Absceso Hepático/cirugía , Humanos , Masculino , Persona de Mediana Edad , Perfusión
6.
Carcinogenesis ; 34(7): 1567-74, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23430954

RESUMEN

Growth arrest-specific gene (Gas) 6 is a γ-carboxyglutamic acid domain-containing protein, which shares 43% amino acid identity with protein S. Gas6 has been shown to enhance cancer cell proliferation in vitro. On the other hand, recent studies have demonstrated that Gas6 inhibits toll-like receptor-mediated immune reactions. Immune reactions are known to affect intestinal tumorigenesis. In this study, we investigated how Gas6 contributes to tumorigenesis in the intestine. Administration of recombinant Gas6 weakly, but significantly, enhanced proliferation of intestinal cancer cells (SW480 and HT29), whereas it suppressed the inflammatory responses of Lipopolysaccharide (LPS)-stimulated monocytes (THP-1). Compared with Gas6(+/+) mice, Gas6(-/-) mice exhibited enhanced azoxymethane/dextran sulfate sodium (DSS)-induced tumorigenesis and had a shorter survival. Gas6(-/-) mice also exhibited more severe DSS-induced colitis. DSS-treated Gas6(-/-) mice showed attenuated Socs1/3 messenger RNA expression and enhanced nuclear factor-kappaB activation in the colonic stroma, suggesting that the target of Gas6 is stromal cells. Bone marrow transplantation experiments indicated that both epithelial cells and bone marrow-derived cells are Gas6 sources. Furthermore, the number of intestinal tumors in Apc(Min) Gas6(-/-) mice was higher than that in Apc(Min) Gas6(+/+) mice, resulting in shorter survival. In a group of 62 patients with advanced colorectal cancer, Gas6 immunoreactivity in cancer tissues was positively correlated with prognosis. Thus, we revealed a unique in vivo inhibitory role of Gas6 during the progression of intestinal tumors associated with suppression of stromal immune reactions. These results suggest a novel therapeutic approach for colorectal cancer patients by regulation of stromal immune responses.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Azoximetano/efectos adversos , Médula Ósea/metabolismo , Trasplante de Médula Ósea/métodos , Proliferación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/patología , Neoplasias Colorrectales/metabolismo , Sulfato de Dextran/farmacología , Progresión de la Enfermedad , Femenino , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monocitos/efectos de los fármacos , FN-kappa B/genética , FN-kappa B/metabolismo , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
7.
Intern Med ; 62(12): 1775-1779, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-36288990

RESUMEN

A 71-year-old woman with recurring stage IV hepatocellular carcinoma (HCC) was admitted to our hospital while being treated with atezolizumab and bevacizumab and complained of fatigue, vomiting, and appetite loss. The following were noted on admission: serum glucose level, 633 mg/dL; metabolic acidemia (HCO3- of 19.5 mmol/L); remarkably low serum and urinary C-peptide levels (0.16 ng/mL and ≤1.5 µg/day, respectively); and urinary ketone body level, 4,197 µmol/L. She was diagnosed with atezolizumab-induced fulminant type 1 diabetes mellitus (T1DM), and insulin therapy improved the symptoms. To our knowledge, this a novel report of atezolizumab-induced fulminant T1DM in an HCC patient.


Asunto(s)
Carcinoma Hepatocelular , Diabetes Mellitus Tipo 1 , Neoplasias Hepáticas , Femenino , Humanos , Anciano , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Recurrencia Local de Neoplasia
8.
Elife ; 102021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33393460

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease. Although rigorous efforts identified the presence of 'cancer stem cells (CSCs)' in PDAC and molecular markers for them, stem cell dynamics in vivo have not been clearly demonstrated. Here we focused on Doublecortin-like kinase 1 (Dclk1), known as a CSC marker of PDAC. Using genetic lineage tracing with a dual-recombinase system and live imaging, we showed that Dclk1+ tumor cells continuously provided progeny cells within pancreatic intraepithelial neoplasia, primary and metastatic PDAC, and PDAC-derived spheroids in vivo and in vitro. Furthermore, genes associated with CSC and epithelial mesenchymal transition were enriched in mouse Dclk1+ and human DCLK1-high PDAC cells. Thus, we provided direct functional evidence for the stem cell activity of Dclk1+ cells in vivo, revealing the essential roles of Dclk1+ cells in expansion of pancreatic neoplasia in all progressive stages.


Asunto(s)
Adenocarcinoma/fisiopatología , Carcinoma Ductal Pancreático/fisiopatología , Linaje de la Célula/genética , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/fisiopatología , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/patología , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología
10.
Sci Rep ; 9(1): 15244, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31645712

RESUMEN

Colonic epithelial cells comprise the mucosal barrier, and their dysfunction promotes microbial invasion from the gut lumen and induces the development of intestinal inflammation. The EP4 receptor is known to mediate the protective effect of prostaglandin (PG) E2 in the gastrointestinal tract; however, the exact role of epithelial EP4 in intestinal pathophysiology remains unknown. In the present study, we aimed to investigate the role of epithelial EP4 in maintaining colonic homeostasis by characterizing the intestinal epithelial cell-specific EP4 knockout (EP4 cKO) mice. Mice harboring the epithelial EP4 deletion showed significantly lower colonic crypt depth and lower numbers of secretory cell lineages, as well as impaired epithelial cells in the colon. Interestingly, EP4-deficient colon epithelia showed a higher number of apoptotic cells. Consistent with the defect in mucosal barrier function of colonic epithelia and secretory cell lineages, EP4 cKO colon stroma showed enhanced immune cell infiltration, which was accompanied by increased production of inflammatory cytokines. Furthermore, EP4-deficient colons were susceptible to dextran sulfate sodium (DSS)-induced colitis. Our study is the first to demonstrate that epithelial EP4 loss resulted in potential "inflammatory" status under physiological conditions. These findings provided insights into the crucial role of epithelial PGE2/EP4 axis in maintaining intestinal homeostasis.


Asunto(s)
Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Colon/patología , Mucosa Intestinal/patología , Subtipo EP4 de Receptores de Prostaglandina E/genética , Animales , Apoptosis , Colitis Ulcerosa/inducido químicamente , Colon/ultraestructura , Sulfato de Dextran , Eliminación de Gen , Ratones , Ratones Noqueados
11.
Dig Liver Dis ; 50(12): 1353-1361, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30001952

RESUMEN

BACKGROUND: Accumulating evidence has shown the existence of tumor stem cells with therapeutic potential. Previously, we reported that doublecortin like kinase 1 (Dclk1) marks tumor stem cells but not normal stem cells in the intestine of ApcMin/+ mice, and that Dclk1- and Lgr5-double positive tumor cells are the tumor stem cells of intestinal tumors. AIM: To investigate molecules highly expressed in the Dclk1+ normal intestinal and Dclk1+ tumor cells in ApcMin/+ mice. METHODS: We used microarray analyses to examine the gene expression profile of Dclk1+ cells in both mouse normal intestinal epithelium and ApcMin/+ mouse intestinal tumors. We also performed immunofluorescence analyses. RESULTS: Genes related to microtubules and the actin cytoskeleton (e.g., Rac2), and members of the Src family kinases (i.e., Hck, Lyn, Csk, and Ptpn6) were highly expressed in both Dclk1+ normal intestinal and Dclk1+ tumor cells. Phosphorylated Hck and phosphorylated Lyn were expressed in Lgr5+ cells in the intestinal tumors of Lgr5EGFP-IRES-CreERT2/+; ApcMin/+ mice. CONCLUSION: We revealed factors that are highly expressed in Dclk1+ intestinal tumor cells, which may help to develop cancer stem cell-targeted therapy in future.


Asunto(s)
Mucosa Intestinal/metabolismo , Neoplasias Intestinales/genética , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transcriptoma , Animales , Quinasas Similares a Doblecortina , Ratones , Análisis por Micromatrices , Fosforilación , Proteínas Proto-Oncogénicas c-hck/metabolismo , Receptores Acoplados a Proteínas G/genética , Familia-src Quinasas/metabolismo
12.
J Clin Invest ; 128(8): 3475-3489, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010625

RESUMEN

Chromatin remodeler Brahma related gene 1 (BRG1) is silenced in approximately 10% of human pancreatic ductal adenocarcinomas (PDAs). We previously showed that BRG1 inhibits the formation of intraductal pancreatic mucinous neoplasm (IPMN) and that IPMN-derived PDA originated from ductal cells. However, the role of BRG1 in pancreatic intraepithelial neoplasia-derived (PanIN-derived) PDA that originated from acinar cells remains elusive. Here, we found that exclusive elimination of Brg1 in acinar cells of Ptf1a-CreER; KrasG12D; Brg1fl/fl mice impaired the formation of acinar-to-ductal metaplasia (ADM) and PanIN independently of p53 mutation, while PDA formation was inhibited in the presence of p53 mutation. BRG1 bound to regions of the Sox9 promoter to regulate its expression and was critical for recruitment of upstream regulators, including PDX1, to the Sox9 promoter and enhancer in acinar cells. SOX9 expression was downregulated in BRG1-depleted ADMs/PanINs. Notably, Sox9 overexpression canceled this PanIN-attenuated phenotype in KBC mice. Furthermore, Brg1 deletion in established PanIN by using a dual recombinase system resulted in regression of the lesions in mice. Finally, BRG1 expression correlated with SOX9 expression in human PDAs. In summary, BRG1 is critical for PanIN initiation and progression through positive regulation of SOX9. Thus, the BRG1/SOX9 axis is a potential target for PanIN-derived PDA.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Transformación Celular Neoplásica/metabolismo , ADN Helicasas/biosíntesis , Proteínas Nucleares/biosíntesis , Neoplasias Pancreáticas/metabolismo , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Factores de Transcripción/biosíntesis , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , ADN Helicasas/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Elementos de Respuesta , Factor de Transcripción SOX9/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA