Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Hepatology ; 77(2): 443-455, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35603471

RESUMEN

BACKGROUND AND AIMS: The mechanism underlying liver regeneration following partial hepatectomy (PH) is not fully elucidated. We aimed to characterize collagen gene expressing hepatic cells following PH and examine their contribution to liver regeneration. APPROACH AND RESULTS: Col-GFP mice, which express GFP under the control of the collagen gene promoter, were used to detect collagen gene expressing cells following PH. The GFP-expressing cells were analyzed via single-cell RNA sequencing (scRNA-seq). Additionally, Col-ER Cre/RFP and Col-ER Cre/DTA mice were utilized to examine the cell fates and functional roles of collagen gene expressing cells in liver regeneration, respectively. The number of collagen gene expressing cells was found to be increased on day 3 and subsequently decreased on day 7 following PH. ScRNA-seq analysis of sorted collagen gene expressing cells showed that the regenerating liver was characterized by three distinct hepatic stellate cell (HSC) clusters, including one representing classic myofibroblasts. The other HSC clusters included an intermediately activated HSC cluster and a proliferating HSC cluster. Of these, the latter cluster was absent in the CCl 4 -induced liver fibrosis model. Cell fate tracing analysis using Col-ER Cre/RFP mice demonstrated that the collagen gene expressing cells escaped death during regeneration and remained in an inactivated state in the liver. Further, depletion of these cells using Col-ER Cre/DTA mice resulted in impaired liver regeneration. CONCLUSIONS: Heterogeneous HSC clusters, one of which was a unique proliferating cluster, were found to appear in the liver following PH. Collagen gene expressing cells, including HSCs, were found to promote liver regeneration.


Asunto(s)
Hepatectomía , Hepatocitos , Ratones , Animales , Hepatocitos/metabolismo , Hígado/metabolismo , Cirrosis Hepática/patología , Células Estrelladas Hepáticas/metabolismo , Colágeno/metabolismo
2.
Gastroenterology ; 156(4): 1140-1155.e4, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30508510

RESUMEN

BACKGROUND & AIMS: Gastritis is associated with development of stomach cancer, but little is known about changes in microRNA expression patterns during gastric inflammation. Specific changes in gene expression in epithelial cells are difficult to monitor because of the heterogeneity of the tissue. We investigated epithelial cell-specific changes in microRNA expression during gastric inflammation and gastritis-associated carcinogenesis in mice. METHODS: We used laser microdissection to enrich epithelial cells from K19-C2mE transgenic mice, which spontaneously develop gastritis-associated hyperplasia, and Gan mice, which express activated prostaglandin E2 and Wnt in the gastric mucosa and develop gastric tumors. We measured expression of epithelial cell-enriched microRNAs and used bioinformatics analyses to integrate data from different systems to identify inflammation-associated microRNAs. We validated our findings in gastric tissues from mice and evaluated protein functions in gastric cell lines (SNU-719, SNU-601, SNU-638, AGS, and GIF-14) and knockout mice. Organoids were cultured from gastric corpus tissues of wild-type and miR-135b-knockout C57BL/6 mice. We measured levels of microRNAs in pairs of gastric tumors and nontumor mucosa from 28 patients in Japan. RESULTS: We found microRNA 135b (miR-135B) to be the most overexpressed microRNA in gastric tissues from K19-C2mE and Gan mice: levels increased during the early stages of gastritis-associated carcinogenesis. Levels of miR-135B were also increased in gastric tumor tissues from gp130F/F mice and patients compared with nontumor tissues. In gastric organoids and immortalized cell lines, expression of miR-135B was induced by interleukin 1 signaling. K19-C2mE mice with disruption of Mir-135b developed hyperplastic lesions that were 50% smaller than mice without Mir-135b disruption and had significant reductions in cell proliferation. Expression of miR-135B in gastric cancer cell lines increased their colony formation, migration, and sphere formation. We identified FOXN3 and RECK messenger RNAs (mRNAs) as targets of miR-135B; their knockdown reduced migration of gastric cancer cell lines. Levels of FOXN3 and RECK mRNAs correlated inversely with levels of miR-135B in human gastric tumors and in inflamed mucosa from K19-C2mE mice. CONCLUSIONS: We found expression of miR-135B to be up-regulated by interleukin L1 signaling in gastric cancer cells and organoids. miR-135B promotes invasiveness and stem-cell features of gastric cancer cells in culture by reducing FOXN3 and RECK messenger RNAs. Levels of these messenger RNA targets, which encode tumor suppressor, are reduced in human gastric tumors.


Asunto(s)
Carcinogénesis/genética , Mucosa Gástrica/patología , Gastritis/genética , Interleucina-1/metabolismo , MicroARNs/genética , Neoplasias Gástricas/genética , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Factores de Transcripción Forkhead , Proteínas Ligadas a GPI/genética , Gastritis/complicaciones , Técnicas de Silenciamiento del Gen , Humanos , Hiperplasia/genética , Ratones , MicroARNs/metabolismo , Organoides/metabolismo , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Neoplasias Gástricas/metabolismo , Regulación hacia Arriba
3.
EMBO Rep ; 19(11)2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30213795

RESUMEN

Glioblastoma is one of the most aggressive forms of cancers and has a poor prognosis. Genomewide analyses have revealed that a set of core signaling pathways, the p53, RB, and RTK pathways, are commonly deregulated in glioblastomas. However, the molecular mechanisms underlying the tumorigenicity of glioblastoma are not fully understood. Here, we show that the lysine deacetylase SIRT2 is required for the proliferation and tumorigenicity of glioblastoma cells, including glioblastoma stem cells. Furthermore, we demonstrate that SIRT2 regulates p73 transcriptional activity by deacetylation of its C-terminal lysine residues. Our results suggest that SIRT2-mediated inactivation of p73 is critical for the proliferation and tumorigenicity of glioblastoma cells and that SIRT2 may be a promising molecular target for the therapy of glioblastoma.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Sirtuina 2/metabolismo , Proteína Tumoral p73/metabolismo , Acetilación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Furanos/farmacología , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Humanos , Lisina/metabolismo , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Quinolinas/farmacología , Sirtuina 2/antagonistas & inhibidores , Sirtuina 2/genética , Células Tumorales Cultivadas , Proteína Tumoral p73/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Sci ; 107(4): 391-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27079437

RESUMEN

Cyclooxygenase-2 (COX-2) and its downstream product prostaglandin E2 (PGE2 ) play a key role in generation of the inflammatory microenvironment in tumor tissues. Gastric cancer is closely associated with Helicobacter pylori infection, which stimulates innate immune responses through Toll-like receptors (TLRs), inducing COX-2/PGE2 pathway through nuclear factor-κB activation. A pathway analysis of human gastric cancer shows that both the COX-2 pathway and Wnt/ß-catenin signaling are significantly activated in tubular-type gastric cancer, and basal levels of these pathways are also increased in other types of gastric cancer. Expression of interleukin-11, chemokine (C-X-C motif) ligand 1 (CXCL1), CXCL2, and CXCL5, which play tumor-promoting roles through a variety of mechanisms, is induced in a COX-2/PGE2 pathway-dependent manner in both human and mouse gastric tumors. Moreover, the COX-2/PGE2 pathway plays an important role in the maintenance of stemness with expression of stem cell markers, including CD44, Prom1, and Sox9, which are induced in both gastritis and gastric tumors through a COX-2/PGE2 -dependent mechanism. In contrast, disruption of Myd88 results in suppression of the inflammatory microenvironment in gastric tumors even when the COX-2/PGE2 pathway is activated, indicating that the interplay of the COX-2/PGE2 and TLR/MyD88 pathways is needed for inflammatory response in tumor tissues. Furthermore, TLR2/MyD88 signaling plays a role in maintenance of stemness in normal stem cells as well as gastric tumor cells. Accordingly, these results suggest that targeting the COX-2/PGE2 pathway together with TLR/MyD88 signaling, which would suppress the inflammatory microenvironment and maintenance of stemness, could be an effective preventive or therapeutic strategy for gastric cancer.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias Gástricas/genética , Receptor Toll-Like 2/genética , Animales , Ciclooxigenasa 2/genética , Dinoprostona/genética , Humanos , Inflamación/genética , Inflamación/patología , Ratones , Factor 88 de Diferenciación Mieloide/genética , Proteínas de Neoplasias/biosíntesis , Transducción de Señal , Neoplasias Gástricas/patología , Receptor Toll-Like 2/metabolismo , Microambiente Tumoral/genética
5.
Biochem Biophys Res Commun ; 444(1): 13-8, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24406169

RESUMEN

Protocadherin10 (PCDH10)/OL-protocadherin is a cadherin-related transmembrane protein that has multiple roles in the brain, including facilitating specific cell-cell connections, cell migration and axon guidance. It has recently been reported that PCDH10 functions as a tumor suppressor and that its overexpression inhibits proliferation or invasion of multiple tumor cells. However, the function of PCDH10 in glioblastoma cells has not been elucidated. In contrast to previous reports on other tumors, we show here that suppression of the expression of PCDH10 by RNA interference (RNAi) induces the growth arrest and apoptosis of glioblastoma cells in vitro. Furthermore, we demonstrate that knockdown of PCDH10 inhibits the growth of glioblastoma cells xenografted into immunocompromised mice. These results suggest that PCDH10 is required for the proliferation and tumorigenicity of glioblastoma cells. We speculate that PCDH10 may be a promising target for the therapy of glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Cadherinas/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Animales , Apoptosis , Neoplasias Encefálicas/genética , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Línea Celular Tumoral , Proliferación Celular , Técnicas de Silenciamiento del Gen , Glioblastoma/genética , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Protocadherinas , Interferencia de ARN
6.
J Biol Chem ; 285(2): 1199-207, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19897489

RESUMEN

Mutation of the tumor suppressor adenomatous polyposis coli (APC) is a key early event in the development of most colorectal tumors. APC promotes degradation of beta-catenin and thereby negatively regulates Wnt signaling, whereas mutated APCs present in colorectal tumor cells are defective in this activity. APC also stimulates the activity of the guanine nucleotide exchange factor Asef and regulates cell morphology and migration. Truncated mutant APCs constitutively activate Asef and induce aberrant migration of colorectal tumor cells. Furthermore, we have recently found that Asef and APC function downstream of hepatocyte growth factor and phosphatidylinositol 3-kinase. We show here that Asef is required for basic fibroblast growth factor- and vascular endothelial growth factor-induced endothelial cell migration. We further demonstrate that Asef is required for basic fibroblast growth factor- and vascular endothelial growth factor-induced microvessel formation. Furthermore, we show that the growth as well as vascularity of subcutaneously implanted tumors are markedly impaired in Asef(-/-) mice compared with wild-type mice. Thus, Asef plays a critical role in tumor angiogenesis and may be a promising target for cancer chemotherapy.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Movimiento Celular , Neoplasias Colorrectales/mortalidad , Células Endoteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neovascularización Patológica/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Neoplasias Colorrectales/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
7.
Oncogene ; 38(22): 4250-4263, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30700829

RESUMEN

We previously showed that NADPH oxidase organizer 1 (Noxo1), a component of NADPH oxidase 1 (NOX1), is a TNF-α-induced tumor-promoting factor in gastric tumorigenesis. However, the mechanism of NOX1-induced reactive oxygen species (ROS) signaling for the gastric tumorigenesis has not been understood. Here, we showed that expression of NOX1 complex components, including Noxo1, but not other NOX family members was significantly upregulated in both mouse models for gastritis and gastric tumors, which was associated with increased ROS levels. We also found that NF-κB directly regulated NOXO1 expression in TNF-α-stimulated gastric cancer cells, suggesting that inflammation induces NOX1 complex activation through TNF-α/NF-κB pathway. Notably, in situ hybridization indicated that Noxo1 mRNA was detected in proliferating cells of gastritis and gastric tumors, and pharmacological inhibition of NOX activity significantly suppressed the proliferation of MKN45 gastric cancer cells and gastric hyperplasia of K19-C2mE mice. These results suggest that NOX1/ROS signaling has an important role in increased proliferation of stomach epithelial cells in the inflamed mucosa. Moreover, we found that expression of SOX2, a marker of gastric epithelial stem cells, was increased by NOX1/ROS signaling. Furthermore, disruption of Noxo1 in K19-C2mE mice significantly suppressed gastritis-associated metaplastic hyperplasia, a potent preneoplastic lesion, which was associated with decreased number of SOX2-positive cells. These results indicate that inflammation-induced Noxo1 expression is responsible for development of metaplastic hyperplasia in the stomach through an increase in SOX2-expressing undifferentiated epithelial cells. Therefore, inhibition of the NOX1/ROS signaling pathway is a possible strategy for prevention and therapy for gastric cancer development.


Asunto(s)
Carcinogénesis/metabolismo , Células Epiteliales/metabolismo , NADPH Oxidasa 1/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción SOXB1/metabolismo , Neoplasias Gástricas/metabolismo , Estómago/patología , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Células Epiteliales/patología , Regulación de la Expresión Génica/fisiología , Humanos , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Neoplasias Gástricas/patología , Factor de Necrosis Tumoral alfa/metabolismo
8.
Adv Biol Regul ; 68: 39-45, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29428221

RESUMEN

Accumulating evidence has indicated that the inflammatory response is important for tumor promotion. However, the mechanisms underlying the induction of the inflammatory response in cancer tissues and how it promotes tumorigenesis remain poorly understood. We constructed several mouse models that develop inflammation-associated gastric and intestinal tumors and examined the in vivo mechanisms of tumorigenesis. Of note, the activation of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway and Toll-like receptor (TLR)/MyD88 signaling cooperatively induced the generation of an inflammatory microenvironment, which is required for early-stage tumorigenesis. The inflammatory response in the stroma induces TNF-α signaling in tumor cells, and the NOX1/ROS signaling pathway is activated downstream. In addition, the inflammatory pathway induces the expression of TLR2 in tumor epithelial cells. Both the NOX1/ROS and TLR2 pathways in tumor cells contribute to the acquisition and maintenance of stemness, which is an important tumor-promoting mechanism stimulated by inflammation. We also found that inflammation promotes malignant processes, like submucosal invasion, of TGF-ß signaling-suppressed tumor cells through the activation of MMP2 protease. Moreover, we showed that mutant p53 induces innate immune and inflammatory signaling in the tumor stroma by a gain-of-function mechanism of mutant p53, which may explain the "cancer-induced inflammation" mechanism. These results indicate that the regulation of the inflammatory microenvironment via the inhibition of the COX-2/PGE2 and TLR/MyD88 pathways in combination will be an effective preventive or therapeutic strategy against gastrointestinal cancer development and malignant progression, especially those carrying p53 gain-of-function mutations.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias Gastrointestinales/metabolismo , Transducción de Señal/fisiología , Animales , Ciclooxigenasa 2/metabolismo , Humanos , Inmunidad Innata/fisiología
9.
Cancer Prev Res (Phila) ; 9(3): 253-63, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26888865

RESUMEN

It has been established that COX-2 and downstream signaling by prostaglandin E2 (PGE2) play a key role in tumorigenesis through generation of inflammatory microenvironment. Toll-like receptor (TLR) signaling through myeloid differentiation factor 88 (MyD88) also regulates inflammatory responses in tumors. However, the relationship between these distinct pathways in tumorigenesis is not yet fully understood. We herein investigated the role of MyD88 in gastric tumorigenesis using Gan mice, which develop inflammation-associated gastric tumors due to the simultaneous activation of the COX-2/PGE2 pathway and Wnt signaling. Notably, the disruption of Myd88 in Gan mice resulted in the significant suppression of gastric tumorigenesis with the inhibition of inflammatory responses, even though COX-2/PGE2 pathway is constitutively activated. Moreover, Myd88 disruption in bone marrow-derived cells (BMDCs) in Gan mice also suppressed inflammation and tumorigenesis, indicating that MyD88 signaling in BMDCs regulates the inflammatory microenvironment. We also found that expression of Tlr2 and its coreceptor Cd14 was induced in tumor epithelial cells in Gan mice, which was suppressed by the disruption of Myd88. It has already been shown that TLR2/CD14 signaling is important for stemness of intestinal epithelial cells. These results indicate that MyD88 in BMDCs, together with COX-2/PGE2 pathway, plays an essential role in the generation of the inflammatory microenvironment, which may promote tumorigenesis through induction of TLR2/CD14 pathway in tumor epithelial cells. These results suggest that inhibition of TLR/MyD88 signaling together with COX-2/PGE2 pathway will be an effective preventive strategy for gastric cancer.


Asunto(s)
Médula Ósea/patología , Transformación Celular Neoplásica/patología , Inflamación/patología , Factor 88 de Diferenciación Mieloide/fisiología , Lesiones Precancerosas/patología , Neoplasias Gástricas/patología , Animales , Apoptosis , Western Blotting , Médula Ósea/metabolismo , Trasplante de Médula Ósea , Proliferación Celular , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Humanos , Técnicas para Inmunoenzimas , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Noqueados , Lesiones Precancerosas/inmunología , Lesiones Precancerosas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Receptor Toll-Like 2/metabolismo , Carga Tumoral , Células Tumorales Cultivadas
10.
J Biol Chem ; 284(33): 22436-22443, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19525225

RESUMEN

Mutations of the tumor suppressor adenomatous polyposis coli (APC) are responsible for sporadic and familial colorectal tumors. APC negatively regulates Wnt signaling by inducing beta-catenin degradation. It has also been shown that APC plays a role in the organization of cytoskeletal networks. APC interacts with Asef and Asef2, Rac1- and Cdc42-specific guanine nucleotide exchange factors (GEFs), and stimulates their GEF activity; thereby regulating cell morphology, adhesion, and migration. Truncated mutant APCs present in colorectal tumor cells activate Asef and Asef2 constitutively and contribute to their aberrant migratory properties. We show here that hepatocyte growth factor (HGF), as well as basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF), induce the accumulation and colocalization of APC and Asef in membrane ruffles and lamellipodia of epithelial cells. Both APC and Asef were found to be required for HGF-induced cell migration. Furthermore, we show that the effects of HGF, bFGF, and EGF on APC and Asef are mediated by the activation of phosphatidylinositol 3-kinase (PI3-kinase) and require the PH domain of Asef. These results suggest that Asef and APC function downstream of HGF and PI3-kinase, and play critical roles in growth factor-mediated regulation of cell morphology and migration.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células CACO-2 , Movimiento Celular , Citoesqueleto/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células HeLa , Humanos , Microscopía Fluorescente/métodos , Modelos Biológicos , Mutación , Estructura Terciaria de Proteína , Factores de Intercambio de Guanina Nucleótido Rho
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA