Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cancer Cell Int ; 18: 156, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30337838

RESUMEN

BACKGROUND: As newly identified Wnt enhancer, R-spondin gene family members have been linked to various cancers; however, their role in isocitrate dehydrogenase-wildtype subtype of human glioblastoma (GBM) cells remains unknown. METHODS: Human U87 and U251 cell lines were used to perform the experiments. GBM stem-like cells were enriched in stem cell growth media and induced to differentiate using retinoid acid or growth factor deprivation. Wnthigh and Wntlow subpopulations were isolated and evaluated by MTS, sphere formation, transwell migration and xenograft formation assays. RESULTS: R-spondin 2 but not R-spondin 3 potentiates Wnt/ß-catenin signaling in GBM cell lines. While R-spondin 2 does not affect cell growth, it induces the expression of pluripotent stem cell markers in combination with Wnt3A. GBM stem-like cells are endowed with intrinsic high activity of ß-catenin signaling, which can be further intensified by R-spondin 2. In addition, R-spondin2 promotes stem cell self-renewal and suppresses retinoid acid- or growth factor deprivation-induced differentiation, indicating R-spondin 2 maintains stem cell traits in GBM. On the other hand, we identify subpopulations of GBM cells that show distinctive responsiveness to Wnt/ß-catenin signaling. Interestingly, Wnthigh and Wntlow cells display distinctive biologic properties. Moreover, Wnthigh cell-inoculated xenografts exhibit enhanced tumorigenicity and increased expression levels of R-spondin 2 compared to Wntlow cell-inoculated xenografts. CONCLUSION: Our study reveals a novel regulatory mechanisms underlying the over-activation of ß-catenin-mediated signaling in the pathogenesis of GBM.

2.
Nitric Oxide ; 34: 27-36, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23669183

RESUMEN

Bleomycin causes acute lung injury through production of reactive species and initiation of inflammation. Previous work has shown alteration to the production of reactive oxygen species results in attenuation of injury. Vitamin E, in particular, γ-tocopherol, isoform, has the potential to scavenge reactive oxygen and nitrogen species. This study examines the utility of dietary supplementation with tocopherols in reducing bleomycin-mediated acute lung injury. Male C57BL6/J mice were intratracheally instilled with PBS or 2 units/kg bleomycin. Animals were analyzed 3 and 8 days post instillation at the cellular, tissue, and organ levels. Results showed successful delivery of tocopherols to the lung via dietary supplementation. Also, increases in reactive oxygen and nitrogen species due to bleomycin are normalized in those mice fed tocopherol diet. Injury was not prevented but inflammation progression was altered, in particular macrophage activation and function. Inflammatory scores based on histology demonstrate limited progression of inflammation in those mice treated with bleomycin and fed tocopherol diet compared to control diet. Upregulation of enzymes and cytokines involved in pro-inflammation were limited by tocopherol supplementation. Day 3 functional changes in elastance in response to bleomycin are prevented, however, 8 days post injury the effect of the tocopherol diet is lost. The effect of tocopherol supplementation upon the inflammatory process is demonstrated by a shift in the phenotype of macrophage activation. The effect of these changes on resolution and the progression of pulmonary fibrosis has yet to be elucidated.


Asunto(s)
Antioxidantes/farmacología , Bleomicina/toxicidad , Pulmón/efectos de los fármacos , Óxido Nítrico/metabolismo , Neumonía/metabolismo , Tocoferoles/farmacología , Administración Oral , Animales , Líquido del Lavado Bronquioalveolar/citología , Ciclooxigenasa 2/metabolismo , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía/tratamiento farmacológico , Neumonía/patología , Especies Reactivas de Oxígeno/metabolismo , Pruebas de Función Respiratoria
3.
Cell Host Microbe ; 31(5): 781-797.e9, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37130518

RESUMEN

Immune checkpoint blockade therapy with anti-PD-1 monoclonal antibody (mAb) is a treatment for colorectal cancer (CRC). However, some patients remain unresponsive to PD-1 blockade. The gut microbiota has been linked to immunotherapy resistance through unclear mechanisms. We found that patients with metastatic CRC who fail to respond to immunotherapy had a greater abundance of Fusobacterium nucleatum and increased succinic acid. Fecal microbiota transfer from responders with low F. nucleatum, but not F. nucleatum-high non-responders, conferred sensitivity to anti-PD-1 mAb in mice. Mechanistically, F. nucleatum-derived succinic acid suppressed the cGAS-interferon-ß pathway, consequently dampening the antitumor response by limiting CD8+ T cell trafficking to the tumor microenvironment (TME) in vivo. Treatment with the antibiotic metronidazole reduced intestinal F. nucleatum abundance, thereby decreasing serum succinic acid levels and resensitizing tumors to immunotherapy in vivo. These findings indicate that F. nucleatum and succinic acid induce tumor resistance to immunotherapy, offering insights into microbiota-metabolite-immune crosstalk in CRC.


Asunto(s)
Neoplasias Colorrectales , Infecciones por Fusobacterium , Animales , Ratones , Fusobacterium nucleatum , Neoplasias Colorrectales/tratamiento farmacológico , Ácido Succínico , Infecciones por Fusobacterium/microbiología , Inmunoterapia , Microambiente Tumoral
4.
Lab Invest ; 90(3): 426-35, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20065943

RESUMEN

The risk of pancreatic cancer is increased in both Snus (the Swedish variant of oral smokeless tobacco) users and, to a greater extent, in cigarette smokers. Concurrent chronic pancreatitis further increases the risk in cigarette smokers. Little is known about the mechanism by which cigarette smoke or Snus increase the risk of pancreatic cancer in individuals with chronic pancreatitis. This study examined the carcinogenic effects of an aqueous extract of cigarette smoke (tobacco smoke, TS) or Snus in an Elastase-IL-1beta transgenic mouse model of chronic pancreatitis. Both transgenic and wild-type (WT) mice were fed diluted TS water or Snus-containing diet for up to 15 months, and monitored for phenotypic and molecular changes in the pancreas. Both TS- and Snus-treated Elastase-IL-1beta mice, but not WT mice, developed significant pancreatic ductal epithelial flattening and severe glandular atrophy compared with untreated transgenic mice. Ductal epithelial cells displayed a high proliferative index, minimal apoptosis, and induction of COX-2 in the setting of chronic inflammation. Up-regulation of TNF-alpha correlated with the onset of severe glandular atrophy. In comparison with Snus-treated mice, TS-Elastase-IL-1beta mice had an earlier onset and a greater extent of phenotypic changes, which were associated with up-regulation of TNF-alpha and increased expression of IL-6, TGF-beta, and SDF-1. Collectively, these findings provide new insights into the mechanism by which tobacco products are likely to promote carcinogenesis in the setting of chronic pancreatitis.


Asunto(s)
Neoplasias Pancreáticas/etiología , Pancreatitis/complicaciones , Fumar/efectos adversos , Tabaco sin Humo/efectos adversos , Animales , Epitelio/patología , Femenino , Fibrosis , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Elastasa Pancreática/genética , Elastasa Pancreática/metabolismo , Pancreatitis/metabolismo , Pancreatitis/patología
5.
Cancer Sci ; 101(2): 559-67, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19922503

RESUMEN

XAF1 (X chromosome-linked inhibitor of apoptosis [XIAP]-associated factor 1) is a novel XIAP modulator that negatively regulates the anti-apoptotic effects of XIAP and sensitizes cells to other cell death triggers. It has been reported to be downregulated in a variety of human cancer cell lines. However, the role of XAF1 in pancreatic carcinogenesis remains unclear. In the present study, we investigated the prognostic values of XAF1 expression and its regulation in cancer cell growth and apoptosis both in vitro and in vivo. From the immunohistochemistry staining of tissue microarray, 40 of 89 (44.9%) pancreatic specimens showed low levels of XAF1 expression. Statistical analysis suggested the downregulation of XAF1 was significantly correlated with tumor staging (P = 0.047) and those patients with low XAF1 levels had shorter survival times (P = 0.0162). Multivariate analysis indicated that XAF1 expression was an independent prognostic indicator of the survival of patients with pancreatic cancer (P = 0.007). Furthermore, we found that restoration of XAF1 expression mediated by Ad5/F35 virus suppressed cell proliferation and induced cell cycle arrest and apoptosis, accompanied by the activation of caspases 3, 8, and 9 and poly(ADP-ribose) polymerase as well as increased level of cytochrome c and Bid cleavage. Notably, XAF1 restoration robustly decreased survivin expression rather than XIAP. In addition, in vivo s.c. xenografts from Ad5/F35-XAF1 treatment, which showed less cellular proliferation and enhanced apoptosis, were significantly smaller than those from control groups. Our findings document that XAF1 is a valuable prognostic marker in pancreatic cancer and could be a potential candidate for cancer gene therapy.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Neoplasias/análisis , Neoplasias Pancreáticas/química , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Femenino , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Persona de Mediana Edad , Proteínas de Neoplasias/fisiología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Pronóstico , Modelos de Riesgos Proporcionales , Proteína Inhibidora de la Apoptosis Ligada a X/análisis , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Lab Invest ; 89(12): 1410-22, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19841619

RESUMEN

Recent studies with Helicobacter-infected mice have shown that bone marrow-derived cells can repopulate the gastric epithelium and progress to cancer. However, it has not been established which cellular subset can potentially contribute to the epithelium. The aim of this study was to investigate the ability of bone marrow-derived mesenchymal stem cells (MSCs) that express cytokeratin 19 (K19) to contribute to the gastric epithelium. MSCs cultures were established from whole bone marrow and expression of K19 was detected in a minority (1 of 13) of clones by real-time PCR and immunostaining. Transfection of a K19-green fluorescent protein (GFP) vector and isolation of GFP-expressing colonies generated high K19-expressing MSC clones (K19GFPMSC). Incubation of MSCs with gastric tissue extract markedly induced mRNA expression of gastric phenotypic markers and was observed to a greater extent in K19GFPMSCs compared with parental MSCs and mock transfectants. Both K19GFPMSCs and GFP-labeled control MSCs gave rise to gastric epithelial cells after injection into the murine stomach. In addition, after blastocyst injections, K19GFPMSCs gave rise to GFP-positive gastric epithelial cells in all 13 pups, whereas only 3 of 10 offspring showed GFP-positive gastric epithelial cells after injection of GFP-labeled control MSCs. Although K19 expression could not be detected in murine whole bone marrow, H. felis infection increased K19-expressing MSCs in the circulation. Taken together, our results show that bone marrow-derived MSCs can contribute to the gastric epithelium. The K19-positive MSC fraction that is induced by chronic H. felis infection appears to be the important subset in this process.


Asunto(s)
Células de la Médula Ósea/fisiología , Diferenciación Celular , Mucosa Gástrica/citología , Queratina-19/metabolismo , Células Madre Mesenquimatosas/fisiología , Animales , Células Sanguíneas , Células de la Médula Ósea/microbiología , Células Cultivadas , Células Clonales/metabolismo , Células Epiteliales/citología , Femenino , Infecciones por Helicobacter/patología , Helicobacter felis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Extractos de Tejidos , Regulación hacia Arriba
7.
Int J Cancer ; 125(3): 688-97, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19358264

RESUMEN

XAF1 (XIAP-associated factor 1) is a novel XIAP binding protein that can antagonize XIAP and sensitize cells to other cell death triggers. Our previous results have shown that aberrant hypermethylation of the CpG sites in XAF1 promoter is strongly associated with lower expression of XAF1 in gastric cancers. In our study, we investigated the effect of restoration of XAF1 expression on growth of gastric cancers. We found that the restoration of XAF1 expression suppressed anchorage-dependent and -independent growth and increased sensitivity to TRAIL and drug-induced apoptosis. Stable cell clones expressing XAF1 exhibited delayed tumor initiation in nude mice. Restoration of XAF1 expression mediated by adenovirus vector greatly increased apoptosis in gastric cancer cell lines in a time- and dose-dependent manner and sensitized cancer cells to TRAIL and drugs-induced apoptosis. Adeno-XAF1 transduction induced cell cycle G2/M arrest and upregulated the expression of p21 and downregulated the expression of cyclin B1 and cdc2. Notably, adeno-XAF1 treatment significantly inhibited tumor growth, strongly enhanced the antitumor activity of TRAIL in a gastric cancer xenograft model in vivo, and significantly prolonged the survival time of animals bearing tumor xenografts. Complete eradication of established tumors was achieved on combined treatment with adeno-XAF1 and TRAIL. Our results document that the restoration of XAF1 inhibits gastric tumorigenesis and tumor growth and that XAF1 is a promising candidate for cancer gene therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Terapia Genética/métodos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/farmacología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/terapia , Proteínas Adaptadoras Transductoras de Señales , Adenoviridae , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Western Blotting , Ciclo Celular/genética , Línea Celular Tumoral , Ciclina B/metabolismo , Ciclina B1 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Humanos , Etiquetado Corte-Fin in Situ , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/uso terapéutico , Plásmidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/fisiopatología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factores de Tiempo , Transducción Genética , Transfección , Trasplante Heterólogo , Regulación hacia Arriba
8.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G385-96, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19541923

RESUMEN

Overexpression of trefoil factor 2 (TFF2) is associated with increased cell migration, resistance to apoptosis, and possibly increased gastric cancer invasion. Dysregulation of p53 is frequently observed in preneoplastic conditions of the stomach. Here, we investigated the effect of p53 on the expression and function of TFF2 in gastric cancer cell lines. Gene expression was determined by reverse transcription-polymerase chain reaction, and promoter activity was assessed by dual luciferase reporter assays. Apoptosis was detected by flow cytometry, and cell migration was evaluated by the Boyden chamber assay. Exogenous expression of p53 dose dependently inhibited endogenous TFF2 mRNA, protein, and promoter activity and resulted in induction of cell apoptosis and inhibition of cell migration. Downregulation of TFF2 by small interfering RNA sensitized gastric cancer cells to drug-induced p53-dependent apoptosis. Addition of human TFF2 peptide reversed p53-dependent apoptosis and inhibition of cell migration. The p53-responsive element was mapped to an AP-1-like cis-element at -182 bp upstream of the TFF2 transcription start site. Mutation of this AP-1-like element abrogated p53-mediated inhibition of TFF2 promoter activity. Gel shift and chromatin immunoprecipitation assays demonstrated that c-Jun and c-Fos bind to this AP-1-like element. Ectopic expression of c-Jun/c-Fos or p300 or treatment of cells with phorbol 12-myristate 13-acetate (PMA) stimulated endogenous TFF2 mRNA expression and promoter activity, and p53 inhibited the effects of AP-1 and PMA on TFF2. p53 induces cell apoptosis and inhibits cell migration in part by downregulating TFF2 expression through an AP-1-like site, suggesting that TFF2 may be an important downstream target of p53.


Asunto(s)
Apoptosis , Movimiento Celular , Mucinas/metabolismo , Proteínas Musculares/metabolismo , Péptidos/metabolismo , Neoplasias Gástricas/metabolismo , Factor de Transcripción AP-1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mucinas/genética , Proteínas Musculares/genética , Mutación , Péptidos/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Interferencia de ARN , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factor de Transcripción AP-1/genética , Transcripción Genética , Transfección , Factor Trefoil-2 , Proteína p53 Supresora de Tumor/genética , Factores de Transcripción p300-CBP/metabolismo
9.
Gastroenterology ; 135(4): 1277-87, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18789941

RESUMEN

BACKGROUND & AIMS: Chronic pancreatitis is a significant cause of morbidity and a known risk factor for pancreatic adenocarcinoma. Interleukin-1beta is a proinflammatory cytokine involved in pancreatic inflammation. We sought to determine whether targeted overexpression of interleukin-1beta in the pancreas could elicit localized inflammatory responses and chronic pancreatitis. METHODS: We created a transgenic mouse model (elastase sshIL-1beta) in which the rat elastase promoter drives the expression of human interleukin-1beta. Mice were followed up for up to 2 years. Pancreata of elastase sshIL-1beta mice were analyzed for chronic pancreatitis-associated histologic and molecular changes. To study the potential effect of p53 mutation in chronic pancreatitis, elastase sshIL-1beta mice were crossed with p53(R172H) mice. RESULTS: Three transgenic lines were generated, and in each line the pancreas was atrophic and occasionally showed dilation of pancreatic and biliary ducts secondary to proximal fibrotic stenosis. Pancreatic histology showed typical features of chronic pancreatitis. There was evidence for increased acinar proliferation and apoptosis, along with prominent expression of tumor necrosis factor-alpha; chemokine (C-X-C motif) ligand 1; stromal cell-derived factor 1; transforming growth factor-beta1; matrix metallopeptidase 2, 7, and 9; inhibitor of metalloproteinase 1; and cyclooxygenase 2. The severity of the lesions correlated well with the level of human interleukin-1beta expression. Older mice displayed acinar-ductal metaplasia but did not develop mouse pancreatic intraepithelial neoplasia or tumors. Elastase sshIL-1beta*p53(R172H/+) mice had increased frequency of tubular complexes, some of which were acinar-ductal metaplasia. CONCLUSIONS: Overexpression of interleukin-1beta in the murine pancreas induces chronic pancreatitis. Elastase sshIL-1beta mice consistently develop severe chronic pancreatitis and constitute a promising model for studying chronic pancreatitis and its relationship with pancreatic adenocarcinoma.


Asunto(s)
Modelos Animales de Enfermedad , Interleucina-1beta/genética , Ratones Transgénicos , Páncreas/fisiopatología , Pancreatitis Crónica/fisiopatología , Animales , Femenino , Fibrosis , Expresión Génica/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Páncreas/patología , Elastasa Pancreática/genética , Pancreatitis Crónica/genética , Pancreatitis Crónica/patología , Fenotipo , Regiones Promotoras Genéticas/genética , Ratas , Índice de Severidad de la Enfermedad
10.
Cancer Res ; 64(16): 5787-94, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15313921

RESUMEN

Protein kinase C (PKC) family, which functions through serine/threonine kinase activity, is involved in signal transduction pathways necessary for cell proliferation, differentiation, and apoptosis. Its critical role in neoplastic transformation and tumor invasion renders PKC a potential target for anticancer therapy. In this study, we investigated the effect of targeting individual PKCs on gastric carcinogenesis. We established gastric cancer cell lines stably expressing antisense PKCalpha, PKCbeta1, and PKCbeta2 cDNA. These stable transfectants were characterized by cell morphology, cell growth, apoptosis, and tumorigenicity in vitro and in vivo. PKCalpha-AS and PKCbeta1-AS transfectants showed a different morphology with flattened, long processes and decreased nuclear:cytoplasmic ratio compared with the control cells. Cell growth was markedly inhibited in PKCalpha-AS and PKCbeta1-AS transfectants. PKCalpha-AS and PKCbeta1-AS cells were more responsive to mitomycin C- or 5-fluorouracil-induced apoptosis. However, antisense targeting of PKCbeta2 did not have any significant effect on cell morphology, cell growth, or apoptosis. Furthermore, antisense inhibition of PKCalpha and PKCbeta1 markedly suppressed colony-forming efficiency in soft agar and in nude mice xenografts. Inhibition of PKCalpha or PKCbeta1 significantly suppressed transcriptional and DNA binding activity of activator protein in gastric cancer cells, suggesting that PKCalpha or PKCbeta1 exerts their effects on cell growth through regulation of activator protein activity. These data provide evidence that targeting PKCalpha and PKCbeta1 by antisense method is a promising therapy for gastric cancer.


Asunto(s)
ADN sin Sentido/administración & dosificación , Proteína Quinasa C/antagonistas & inhibidores , Neoplasias Gástricas/terapia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Adhesión Celular/genética , División Celular/genética , Línea Celular Tumoral , ADN sin Sentido/genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Quinasa C/genética , Proteína Quinasa C beta , Proteína Quinasa C-alfa , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factor de Transcripción AP-1/antagonistas & inhibidores , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Res ; 63(22): 7724-32, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14633697

RESUMEN

Survivin plays an important role in cancer development. We aim to show here that suppression of survivin expression or function by antisense and dominant-negative (DN) mutant can inhibit gastric cancer carcinogenesis and angiogenesis in vivo. Plasmid constructs expressing survivin antisense and DN mutant replacing the cysteine residue at amino acid 84 with alanine (Cys84Ala) were prepared and introduced into BCG-823 and MKN-45 gastric cancer cells to establish stable transfectants. We showed that both antisense and DN mutant stable transfectants exhibited abnormal morphology, with decreased cell growth and increased rate of spontaneous apoptosis and mitotic catastrophe. Furthermore, in nude mice xenografts, these cells exhibited decreased de novo gastric tumor formation and reduced development of angiogenesis. Results from these studies strongly suggest that survivin is a promising target for gastric cancer treatment.


Asunto(s)
Adenocarcinoma/terapia , ADN sin Sentido/genética , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Neovascularización Patológica/terapia , Neoplasias Gástricas/terapia , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis/genética , División Celular/genética , Línea Celular Tumoral , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas de Neoplasias , Neovascularización Patológica/genética , Neoplasias Gástricas/irrigación sanguínea , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Survivin , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Gastrointest Tumors ; 3(1): 25-36, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27722154

RESUMEN

BACKGROUND: Gastric cancer is a heterogeneous malignant disease associated with environmental and genetic predisposing factors. While gastric cancer incidence and mortality fell greatly globally over the past decades, it remains the fourth cause of cancer-related death worldwide. Thus, prevention of gastric cancer is still a major strategy for improvement of gastric cancer prognosis. SUMMARY: Helicobacter pylori infection has been demonstrated to be a major risk factor for the development of gastric cancer. Unhealthy diet and lifestyle, including high-salt food, smoking and drinking, are able to induce genotypic and phenotypic transformation of gastric epithelial cells. Gene mutations (such as E-cadherin) in stomach epithelial cells are major genetic causes for gastric cancer. The eradication of H. pylori has been demonstrated to be an effective approach for primary prevention of gastric cancer. Increased intake of a diet rich in vegetables and fresh fruits as well as smoking cessation have been shown to reduce the incidence of gastric cancer. The secondary prevention strategy is to screen premalignant gastric lesions by endoscopy. Biomarker tests are also reliable methods to identify gastric precancerous lesions. Endoscopy screening is still the gold standard for diagnosis of gastric cancer. KEY MESSAGE: H. pylori infection, a diet rich in salted and/or smoked food and red meat, as well as gene mutations are major risk factors for the development of gastric cancer. PRACTICAL IMPLICATIONS: The eradication of H. pylori is a major primary preventive strategy of gastric cancer. A healthy lifestyle, including increased intake of a diet rich in fruit and vegetables, reduced intake of salted and smoked food and red meat, a reduction of alcohol intake as well as smoking cessation will be effective approaches for the prevention of gastric cancer.

13.
Oncotarget ; 7(6): 7096-109, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26771139

RESUMEN

Survivin overexpression is associated with poor prognosis of human gastric cancer, and is a target for gastric cancer therapy. YM155 is originally identified as a specific inhibitor of survivin. In this study, we investigated the antitumor effect of YM155 on human gastric cancer. Our results showed that YM155 treatment significantly inhibited cell proliferation, reduced colony formation and induced apoptosis of gastric cancer cells in a dose-dependent manner. Accordingly, YM155 treatment significantly decreased survivin expression without affecting XIAP expression and increased the cleavage of apoptosis-associated proteins caspase 3, 7, 8, 9. YM155 significantly inhibited sphere formation of gastric cancer cells, suppressed expansion and growth of the formed spheres (cancer stem cell-like cells, CSCs) and downregulated the protein levels of ß-catenin, c-Myc, Cyclin D1 and CD44 in gastric cancer cells. YM155 infusion at 5 mg/kg/day for 7 days markedly inhibited growth of gastric cancer xenograft in a nude mouse model. Immunohistochemistry staining and Western Blot showed that YM155 treatment inhibited expression of survivin and CD44, induced apoptosis and reduced CD44+ CSCs in xenograft tumor tissues in vivo. No obvious pathological changes were observed in organs (e.g. heart, liver, lung and kidney) in YM155-treated mice. Our results demonstrated that YM155 inhibits cell proliferation, induces cell apoptosis, reduces cancer stem cell expansion, and inhibits xenograft tumor growth in gastric cancer cells. Our results elucidate a new mechanism by which YM155 inhibits gastric cancer growth by inhibition of CSCs. YM155 may be a promising agent for gastric cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Mucosa Gástrica/metabolismo , Imidazoles/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Naftoquinonas/farmacología , Neoplasias Gástricas/patología , Neoplasias Gástricas/prevención & control , Animales , Western Blotting , Femenino , Humanos , Técnicas para Inmunoenzimas , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estómago/patología , Neoplasias Gástricas/metabolismo , Survivin , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Oncogene ; 35(41): 5388-5399, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27109105

RESUMEN

Bone marrow-derived cells have important roles in cancer development and progression. Our previous studies demonstrated that murine bone marrow-derived myofibroblasts (BMFs) enhanced tumor growth. In this study, we investigated the mechanisms of BMF actions. We found that co-injection of BMFs with gastric cancer cells markedly promoted tumorigenesis. Co-cultured BMFs or BMF-conditioned medium (BMF-CM) induced the formation of spheres, which expressed stem cell signatures and exhibited features of self-renewal, epithelial-to-mesenchymal transition and tumor initiation. Furthermore, CD44+ fractions in spheres were able to initiate tumorigenesis and re-establish tumors in serially passaged xenografts. In co-culture systems, BMFs secreted high levels of murine interleukin-6 (IL-6) and hepatocyte growth factor (HGF), whereas cancer cells produced high level of transformation growth factor-ß1 (TGF-ß1). BMF-CM and IL-6 activated BMFs to produce mHGF, which activated signal transducer and activator of transcription 3 (STAT3) and upregulated TGF-ß1 in human cancer cells. In return, cancer cell-CM stimulated BMFs to produce IL-6, which was inhibited by anti-TGF-ß1 neutralizing antibody. Blockade of HGF/Met, Janus kinase 2 (JAK2)/STAT3 and TGF-ß1 signaling by specific inhibitors inhibited BMF-induced sphere formation. STAT3 knockdown in cancer cells also inhibited BMF-induced sphere formation and tumorigenesis. Moreover, TGF-ß1 overexpression in cancer cells was co-related with IL-6 and HGF overexpression in stromal cells in human gastric cancer tissues. Our results show that BMF-derived IL-6/HGF and cancer cell-derived TGF-ß1 mediate the interactions between BMFs and gastric cancer cells, which regulate cancer stemness and promote tumorigenesis. Targeting inhibition of the interactions between BMFs and cancer cells may be a new strategy for cancer therapy.


Asunto(s)
Carcinogénesis/genética , Factor de Crecimiento de Hepatocito/genética , Interleucina-6/genética , Neoplasias Gástricas/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Transición Epitelial-Mesenquimal/genética , Humanos , Janus Quinasa 2/genética , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Células Madre Neoplásicas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
15.
Zhonghua Yi Xue Za Zhi ; 84(24): 2064-9, 2004 Dec 17.
Artículo en Zh | MEDLINE | ID: mdl-15730617

RESUMEN

OBJECTIVE: To investigate the activation of signal transducers and activators of transcription 3 (Stat3) in different types of gastric cancer cell lines and tissues and evaluate the relationship with their clinicopathological parameters. METHODS: Western blotting and electrophoretic mobility shift assay (EMSA) were used to detected the expression of Stat3 protein and Stat3 DNA-binding activity in normal human gastric epithelial cell line 3T3 and five gastric cancer cell lines with different differentiation: MKN28, SGC7901, MKN45, AGS and NCI-SNU-1, respectively. The localization of phospho-Stat3 was determined by immunocytochemistry. The expressive intensity of phospho-Stat3 protein in 50 cases of gastric cancer tissues and adjacent normal mucosa were measured by immunohistochemistry. RESULTS: Compared with normal gastric epithelial cell line 3T3, elevated activities of Stat3 were found in five different human gastric cancer cell lines. The Stat3 DNA-binding activity in moderately and poorly differentiated stomach adenocarcinoma cell lines (SGC7901, MKN45 and AGS) was higher than that of other cell lines (MKN28 and NCI-SNU-1). Phospho-Stat3 was detected primarily in the nuclei of AGS cells. The expressive intensity of phospho-Stat3 protein was significantly increased in gastric cancer tissues as compared with the adjacent normal gastric mucosa, especially in moderately and poorly differentiated cancers (both P < 0.05). The expressive intensity of phospho-Stat3 protein in stage II and stage III tumors was higher than that in stage I tumors (P < 0.05). No statistic difference of phospho-Stat3 expression was found between stage IV and stage I tumors (P > 0.05). The expression of phospho-Stat3 was closely correlated with the differentiation of gastric cancer. CONCLUSION: Elevated activity of Stat3 can be found in different types of human gastric cancer cell lines and gastric cancer. JAK/STAT signal transduction pathway may play an important role in the development of human gastric cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma de Células en Anillo de Sello/metabolismo , Factor de Transcripción STAT3/biosíntesis , Neoplasias Gástricas/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Carcinoma de Células en Anillo de Sello/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Metiltransferasas/biosíntesis , Proteína Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas , Factor de Transcripción STAT3/genética , Transducción de Señal , Neoplasias Gástricas/patología , Transactivadores/metabolismo
16.
Oncotarget ; 5(14): 5403-15, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24980821

RESUMEN

X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1), a XIAP-binding protein, is a tumor suppressor gene. XAF1 was silent or expressed lowly in most human malignant tumors. However, the role of XAF1 in hepatocellular carcinoma (HCC) remains unknown. In this study, we investigated the effect of XAF1 on tumor growth and angiogenesis in hepatocellular cancer cells. Our results showed that XAF1 expression was lower in HCC cell lines SMMC-7721, Hep G2 and BEL-7404 and liver cancer tissues than that in paired non-cancer liver tissues. Adenovirus-mediated XAF1 expression (Ad5/F35-XAF1) significantly inhibited cell proliferation and induced apoptosis in HCC cells in dose- and time- dependent manners. Infection of Ad5/F35-XAF1 induced cleavage of caspase -3, -8, -9 and PARP in HCC cells. Furthermore, Ad5/F35-XAF1 treatment significantly suppressed tumor growth in a xenograft model of liver cancer cells. Western Blot and immunohistochemistry staining showed that Ad5/F35-XAF1 treatment suppressed expression of vascular endothelial growth factor (VEGF), which is associated with tumor angiogenesis, in cancer cells and xenograft tumor tissues. Moreover, Ad5/F35-XAF1 treatment prolonged the survival of tumor-bearing mice. Our results demonstrate that XAF1 inhibits tumor growth by inducing apoptosis and inhibiting tumor angiogenesis. XAF1 may be a promising target for liver cancer treatment.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Proteínas de Neoplasias/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación hacia Abajo , Terapia Genética , Células HEK293 , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/patología , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/terapia , Transfección
17.
Cancer Lett ; 343(1): 80-9, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24145153

RESUMEN

Bone marrow-derived myofibroblasts (BMFs) have been shown to promote tumor growth. Here, we found that BMFs or BMF conditioned medium (BMF-CM) induced cancer stem cell-like sphere formation of colon cancer cells. The co-cultured BMFs, but not co-cultured cancer cells, expressed higher levels of IL-6 than BMFs or cancer cells cultured alone. Anti-mouse IL-6 neutralizing antibody, JAK2 inhibitors and STAT3 knockdown in mouse cancer cells reduced BMF- and BMF-CM-induced sphere formation of colon cancer cells. When co-injected, BMFs significantly enhanced tumorigenesis of colon cancer cells in mice. Our results demonstrate that BMFs promote tumorigenesis via the activation of the IL-6/JAK2/STAT3 pathway.


Asunto(s)
Células de la Médula Ósea/citología , Neoplasias del Colon/patología , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Miofibroblastos/citología , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Técnicas de Cocultivo , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias
18.
Cancer Prev Res (Phila) ; 5(2): 205-15, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22030090

RESUMEN

Myeloid-derived suppressor cells (MDSC) accumulate in the spleen and tumors and contribute to tumor growth, angiogenesis, and progression. In this study, we examined the effects of curcumin on the activation and differentiation of MDSCs, their interaction with human cancer cells, and related tumor growth. Treatment with curcumin in the diet or by intraperitoneal injection significantly inhibited tumorigenicity and tumor growth, decreased the percentages of MDSCs in the spleen, blood, and tumor tissues, reduced interleukin (IL)-6 levels in the serum and tumor tissues in a human gastric cancer xenograft model and a mouse colon cancer allograft model. Curcumin treatment significantly inhibited cell proliferation and colony formation of cancer cells and decreased the secretion of murine IL-6 by MDSCs in a coculture system. Curcumin treatment inhibited the expansion of MDSCs, the activation of Stat3 and NF-κB in MDSCs, and the secretion of IL-6 by MDSCs, when MDSCs were cultured in the presence of IL-1ß, or with cancer cell- or myofibroblast-conditioned medium. Furthermore, curcumin treatment polarized MDSCs toward a M1-like phenotype with an increased expression of CCR7 and decreased expression of dectin 1 in vivo and in vitro. Our results show that curcumin inhibits the accumulation of MDSCs and their interaction with cancer cells and induces the differentiation of MDSCs. The induction of MDSC differentiation and inhibition of the interaction of MDSCs with cancer cells are potential strategies for cancer prevention and therapy.


Asunto(s)
Anticarcinógenos/farmacología , Diferenciación Celular , Neoplasias del Colon/tratamiento farmacológico , Curcumina/farmacología , Células Mieloides/metabolismo , Células Mieloides/patología , Neoplasias Gástricas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Mieloides/efectos de los fármacos , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Oncol Rep ; 25(4): 1039-46, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21279308

RESUMEN

Colon cancer is one of the most common cancers. Survivin is overexpressed in human colon cancer and correlate with chemoresistance, angiogenesis and poor prognosis. Oxaliplatin, a platinum derivative cancer drug, has been used for treating human colorectal cancers. In the present study, we investigated the effect of the adeno-associated virus (AAV)-mediated survivin mutant Thr34Ala [rAAV-Sur-Mut(T34A)] on colon cancer growth. Infection with rAAV-Sur-Mut(T34A) inhibited cell proliferation, induced apoptosis and mitotic catastrophe, and sensitized colon cancer cells to chemotherapeutic drugs in vitro. Treatment with rAAV-Sur-Mut(T34A) significantly induced apoptosis, reduced angiogenesis and inhibited colon cancer growth in vivo. More importantly, rAAV-Sur-Mut(T34A) treatment strongly enhanced the anti-tumor activity of oxaliplatin and prolonged animal survival. Thus, the use of rAAV-Sur-Mut(T34A) in combination with chemotherapy may be a promising strategy for colon cancer therapy.


Asunto(s)
Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Dependovirus/genética , Proteínas Inhibidoras de la Apoptosis/genética , Mutación/genética , Neovascularización Patológica/prevención & control , Compuestos Organoplatinos/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis , Western Blotting , Proliferación Celular , Neoplasias del Colon/prevención & control , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Ratones Endogámicos BALB C , Oxaliplatino , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Survivin , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Lett ; 310(2): 170-80, 2011 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-21788101

RESUMEN

Autophagy is designated as type II programmed cell death and may confer a tumor-suppressive function. Our previous studies have shown that XIAP-associated factor 1 (XAF1) induced apoptosis and inhibited tumor growth in gastric cancer cells. In this study, we investigated the effect of XAF1 on the induction of autophagy in gastric cancer cells. We found that adenovirus vector-mediated XAF1 (adeno-XAF1) expression markedly induced autophagy, upregulated the level of Beclin-1 and inhibited phospho-Akt and phospho-p70S6K in gastric cancer cells. The downregulation of Beclin 1 or 3-methyladenine treatment suppressed adeno-XAF1-induced autophagy, but significantly enhanced adeno-XAF1-induced apoptosis. A pan-caspase inhibitor prevented adeno-XAF1-induced apoptosis, but significantly increased adeno-XAF1-induced autophagy. Furthermore, adeno-XAF1 induced autophagy in xenograft tumor and inhibited tumor growth. Our results document that adeno-XAF1 induces autophagy through upregulation of Beclin 1 expression and inhibition of Akt/p70S6K pathway, and reveal a new mechanism of XAF1 tumor suppression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Autofagia/genética , Proteínas de la Membrana/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Neoplasias Gástricas/terapia , Proteínas Adaptadoras Transductoras de Señales , Adenoviridae/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Línea Celular Tumoral , Femenino , Genes Supresores de Tumor , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Transfección , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA