Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Semin Cancer Biol ; 86(Pt 3): 445-456, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35131480

RESUMO

Alterations in metabolic pathways are a hallmark of cancer. A deeper understanding of the contribution of different metabolites to carcinogenesis is thus vitally important to elucidate mechanisms of tumor initiation and progression to inform therapeutic strategies. Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide and its altered metabolic landscape is beginning to unfold with the advancement of technologies. In particular, characterization of the lipidome of human HCCs has accelerated, and together with biochemical analyses, are revealing recurrent patterns of alterations in glycerophospholipid, sphingolipid, cholesterol and bile acid metabolism. These widespread alterations encompass a myriad of lipid species with numerous roles affecting multiple hallmarks of cancer, including aberrant growth signaling, metastasis, evasion of cell death and immunosuppression. In this review, we summarize the current trends and findings of the altered lipidomic landscape of HCC and discuss their potential biological significance for hepatocarcinogenesis.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/metabolismo , Lipidômica , Carcinogênese/genética , Transformação Celular Neoplásica
2.
Amino Acids ; 53(12): 1807-1815, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33646427

RESUMO

Dysregulated cellular energetics has recently been recognized as a hallmark of cancer and garnered attention as a potential targeting strategy for cancer therapeutics. Cancer cells reprogram metabolic activities to meet bio-energetic, biosynthetic and redox requirements needed to sustain indefinite proliferation. In many cases, metabolic reprogramming is the result of complex interactions between genetic alterations in well-known oncogenes and tumor suppressors and epigenetic changes. While the metabolism of the two most abundant nutrients, glucose and glutamine, is reprogrammed in a wide range of cancers, accumulating evidence demonstrates that additional metabolic pathways are also critical for cell survival and growth. Proline metabolism is one such metabolic pathway that promotes tumorigenesis in multiple cancer types, including liver cancer, which is the fourth main cause of cancer mortality in the world. Despite the recent spate of approved treatments, including targeted therapy and combined immunotherapies, there has been no significant gain in clinical benefits in the majority of liver cancer patients. Thus, exploring novel therapeutic strategies and identifying new molecular targets remains a top priority for liver cancer. Two of the enzymes in the proline biosynthetic pathway, pyrroline-5-carboxylate reductase (PYCR1) and Aldehyde Dehydrogenase 18 Family Member A1 (ALDH18A1), are upregulated in liver cancer of both human and animal models, while proline catabolic enzymes, such as proline dehydrogenase (PRODH) are downregulated. Here we review the latest evidence linking proline metabolism to liver and other cancers and potential mechanisms of action for the proline pathway in cancer development.


Assuntos
Carcinogênese/metabolismo , Reprogramação Celular/fisiologia , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Fígado/patologia , Mitocôndrias/metabolismo , Prolina/metabolismo , Animais , Humanos , Neoplasias Hepáticas/patologia , Mitocôndrias/patologia
3.
J Hepatol ; 72(4): 725-735, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31726117

RESUMO

BACKGROUND & AIM: Under the regulation of various oncogenic pathways, cancer cells undergo adaptive metabolic programming to maintain specific metabolic states that support their uncontrolled proliferation. As it has been difficult to directly and effectively inhibit oncogenic signaling cascades with pharmaceutical compounds, focusing on the downstream metabolic pathways that enable indefinite growth may provide therapeutic opportunities. Thus, we sought to characterize metabolic changes in hepatocellular carcinoma (HCC) development and identify metabolic targets required for tumorigenesis. METHODS: We compared gene expression profiles of Morris Hepatoma (MH3924a) and DEN (diethylnitrosamine)-induced HCC models to those of liver tissues from normal and rapidly regenerating liver models, and performed gain- and loss-of-function studies of the identified gene targets for their roles in cancer cell proliferation in vitro and in vivo. RESULTS: The proline biosynthetic enzyme PYCR1 (pyrroline-5-carboxylate reductase 1) was identified as one of the most upregulated genes in the HCC models. Knockdown of PYCR1 potently reduced cell proliferation of multiple HCC cell lines in vitro and tumor growth in vivo. Conversely, overexpression of PYCR1 enhanced the proliferation of the HCC cell lines. Importantly, PYCR1 expression was not elevated in the regenerating liver, and KD or overexpression of PYCR1 had no effect on proliferation of non-cancerous cells. Besides PYCR1, we found that additional proline biosynthetic enzymes, such as ALDH18A1, were upregulated in HCC models and also regulated HCC cell proliferation. Clinical data demonstrated that PYCR1 expression was increased in HCC, correlated with tumor grade, and was an independent predictor of clinical outcome. CONCLUSION: Enhanced expression of proline biosynthetic enzymes promotes HCC cell proliferation. Inhibition of PYCR1 or ALDH18A1 may be a novel therapeutic strategy to target HCC. LAY SUMMARY: Even with the recently approved immunotherapies against liver cancer, currently available medications show limited clinical benefits or efficacy in the majority of patients. As such, it remains a top priority to discover new targets for effective liver cancer treatment. Here, we identify a critical role for the proline biosynthetic pathway in liver cancer development, and demonstrate that targeting key proteins in the pathway, namely PYCR1 and ALDH18A1, may be a novel therapeutic strategy for liver cancer.


Assuntos
Carcinogênese/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas/metabolismo , Prolina/biossíntese , Transdução de Sinais/genética , Aldeído Desidrogenase/deficiência , Aldeído Desidrogenase/genética , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/patologia , Proliferação de Células/genética , Dietilnitrosamina/efeitos adversos , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Células HaCaT , Células Hep G2 , Humanos , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Pirrolina Carboxilato Redutases/deficiência , Pirrolina Carboxilato Redutases/genética , Ratos , Transcriptoma , Transfecção , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto , delta-1-Pirrolina-5-Carboxilato Redutase
4.
Hum Mol Genet ; 23(2): 502-13, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24026679

RESUMO

Seipin regulates lipid homeostasis by preventing lipid droplet (LD) formation in non-adipocytes but promoting it in developing adipocytes. Here, we report that seipin interacts with 14-3-3ß through its N- and C-termini. Expression of 14-3-3ß is upregulated during adipogenesis, and its deletion results in defective adipogenesis without affecting key adipogenic transcription factors. We further identified the actin-severing protein cofilin-1 as an interacting partner to 14-3-3ß. Cofilin-1 was spatiotemporally recruited by 14-3-3ß in the cytoplasm during adipocyte differentiation. Extensive actin cytoskeleton remodelling, from stress fibres to cortical structures, was apparent during adipogenesis, but not under lipogenic conditions, indicating that actin cytoskeleton remodelling is only required for adipocyte development. Similar to seipin and 14-3-3ß, cofilin-1 knockdown led to impaired adipocyte development. At the cellular level, differentiated cells with knockdown of cofilin-1, 14-3-3ß or seipin continued to maintain relatively intact stress fibres, in contrast to cortical actin structure in control cells. Finally, 3T3-L1 cells expressing a severing-resistant actin mutant exhibited impaired adipogenesis. We propose that seipin regulates adipogenesis by recruiting cofilin-1 to remodel actin cytoskeleton through the 14-3-3ß protein.


Assuntos
Proteínas 14-3-3/metabolismo , Citoesqueleto de Actina/metabolismo , Adipócitos/fisiologia , Adipogenia , Cofilina 1/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Células 3T3-L1 , Animais , Subunidades gama da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Proteínas Heterotriméricas de Ligação ao GTP/genética , Humanos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL
5.
EMBO Rep ; 15(6): 714-22, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24711543

RESUMO

While molecular regulation of insulin granule exocytosis is relatively well understood, insulin granule biogenesis and maturation and its influence on glucose homeostasis are relatively unclear. Here, we identify a novel protein highly expressed in insulin-secreting cells and name it BIG3 due to its similarity to BIG/GBF of the Arf-GTP exchange factor (GEF) family. BIG3 is predominantly localized to insulin- and clathrin-positive trans-Golgi network (TGN) compartments. BIG3-deficient insulin-secreting cells display increased insulin content and granule number and elevated insulin secretion upon stimulation. Moreover, BIG3 deficiency results in faster processing of proinsulin to insulin and chromogranin A to ß-granin in ß-cells. BIG3-knockout mice exhibit postprandial hyperinsulinemia, hyperglycemia, impaired glucose tolerance, and insulin resistance. Collectively, these results demonstrate that BIG3 negatively modulates insulin granule biogenesis and insulin secretion and participates in the regulation of systemic glucose homeostasis.


Assuntos
Homeostase/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas/metabolismo , Vesículas Secretórias/química , Animais , Calorimetria Indireta , Glucose/fisiologia , Homeostase/fisiologia , Hiperglicemia/genética , Insulina/análise , Resistência à Insulina/genética , Secreção de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Proteínas/genética , Rede trans-Golgi/metabolismo
6.
Biochem J ; 464(2): 179-92, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25220164

RESUMO

Extensive actin cytoskeleton remodelling occurs during adipocyte development. We have previously shown that disruption of stress fibres by the actin-severing protein cofilin is a requisite step in adipogenesis. However, it remains unclear whether actin nucleation and assembly into the cortical structure are essential for adipocyte development. In the present study we investigated the role of cortical actin assembly and of actin nucleation by the actin-related protein 2/3 (Arp2/3) complex in adipogenesis. Cortical actin structure formation started with accumulation of filamentous actin (F-actin) patches near the plasma membrane during adipogenesis. Depletion of Arp2/3 by knockdown of its subunits Arp3 or ARPC3 strongly impaired adipocyte differentiation, although adipogenesis-initiating factors were unaffected. Moreover, the assembly of F-actin-rich structures at the plasma membrane was suppressed and the cortical actin structure poorly developed after adipogenic induction in Arp2/3-deficient cells. Finally, we provide evidence that the cortical actin cytoskeleton is essential for efficient glucose transporter 4 (GLUT4) vesicle exocytosis and insulin signal transduction. These results show that the Arp2/3 complex is an essential regulator of adipocyte development through control of the formation of cortical actin structures, which may facilitate nutrient uptake and signalling events.


Assuntos
Citoesqueleto de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Proteína 2 Relacionada a Actina/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Adipogenia , Citoesqueleto de Actina/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/química , Adipócitos/metabolismo , Animais , Diferenciação Celular/genética , Transportador de Glucose Tipo 4/metabolismo , Insulina/metabolismo , Camundongos , Transdução de Sinais
7.
Gut ; 63(3): 385-94, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23729675

RESUMO

OBJECTIVE: To investigate the role of obesity-associated inflammation and immune modulation in gastric carcinogenesis during Helicobacter-induced chronic gastric inflammation. DESIGN: C57BL/6 male mice were infected with H felis and placed on a high-fat diet (45% calories from fat). Study animals were analysed for gastric and adipose pathology, inflammatory markers in serum, stomach and adipose tissue, and immune responses in blood, spleen, stomach and adipose tissue. RESULTS: H felis-induced gastric carcinogenesis was accelerated in diet-induced obese mice compared with lean controls. Obesity increased bone marrow-derived immature myeloid cells in blood and gastric tissue of H felis-infected mice. Obesity also led to elevations in CD4 T cells, IL-17A, granulocyte macrophage colony-stimulating factor, phosphorylated STAT3 and prosurvival gene expression in gastric tissue of H felis-infected mice. Conversely, in adipose tissue of obese mice, H felis infection increased macrophage accumulation and expression of IL-6, C-C motif ligand 7 (CCL7) and leptin. Finally, the combination of obesity and gastric inflammation synergistically increased serum proinflammatory cytokines, including IL-6. CONCLUSIONS: Here, we have established a model to study the molecular mechanism by which obesity predisposes individuals to gastric cancer. In H felis-infected mice, obesity increased proinflammatory immune responses and accelerated gastric carcinogenesis. Interestingly, gastric inflammation augmented obesity-induced adipose inflammation and production of adipose-derived factors in obese, but not lean, mice. Our findings suggest that obesity accelerates Helicobacter-associated gastric cancer through cytokine-mediated cross-talk between inflamed gastric and adipose tissues, augmenting immune responses at both tissue sites, and thereby contributing to a protumorigenic gastric microenvironment.


Assuntos
Gastrite/complicações , Infecções por Helicobacter/complicações , Helicobacter felis , Células Progenitoras Mieloides/fisiologia , Obesidade/complicações , Neoplasias Gástricas/etiologia , Células Th17/fisiologia , Animais , Biomarcadores/metabolismo , Movimento Celular , Citocinas/metabolismo , Dieta Hiperlipídica , Citometria de Fluxo , Gastrite/diagnóstico , Gastrite/metabolismo , Gastrite/microbiologia , Infecções por Helicobacter/imunologia , Inflamação/complicações , Inflamação/metabolismo , Inflamação/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/imunologia , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/microbiologia
8.
Lab Invest ; 89(12): 1410-22, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19841619

RESUMO

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.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular , Mucosa Gástrica/citologia , Queratina-19/metabolismo , Células-Tronco Mesenquimais/fisiologia , Animais , Células Sanguíneas , Células da Medula Óssea/microbiologia , Células Cultivadas , Células Clonais/metabolismo , Células Epiteliais/citologia , Feminino , Infecções por Helicobacter/patologia , Helicobacter felis/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Extratos de Tecidos , Regulação para Cima
9.
Mol Cell Oncol ; 6(3): 1585171, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31131306

RESUMO

The mammalian target of rapamycin complex 1 (mTORC1) plays an important role in the development and progression of multiple cancers. Its activity is regulated by both growth factor and nutrient signals, and the branched-chain amino acid (BCAA) leucine plays an important and unique role in this process. Recently we found that cancers of the liver and multiple other tissues suppress the catabolism of BCAAs, thereby facilitating the chronic activation of mTORC1. Our results unveil how mTORC1's nutrient-sensing arm can be manipulated by tumors, and suggest that restoring BCAA catabolism may help control mTORC1 activity in cancer cells.

10.
Cell Metab ; 29(5): 1151-1165.e6, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661928

RESUMO

Tumors display profound changes in cellular metabolism, yet how these changes aid the development and growth of tumors is not fully understood. Here we use a multi-omic approach to examine liver carcinogenesis and regeneration, and find that progressive loss of branched-chain amino acid (BCAA) catabolism promotes tumor development and growth. In human hepatocellular carcinomas and animal models of liver cancer, suppression of BCAA catabolic enzyme expression led to BCAA accumulation in tumors, though this was not observed in regenerating liver tissues. The degree of enzyme suppression strongly correlated with tumor aggressiveness, and was an independent predictor of clinical outcome. Moreover, modulating BCAA accumulation regulated cancer cell proliferation in vitro, and tumor burden and overall survival in vivo. Dietary BCAA intake in humans also correlated with cancer mortality risk. In summary, loss of BCAA catabolism in tumors confers functional advantages, which could be exploited by therapeutic interventions in certain cancers.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Carcinogênese/metabolismo , Carcinoma Hepatocelular/metabolismo , Progressão da Doença , Regulação para Baixo , Neoplasias Hepáticas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Aminoácidos de Cadeia Ramificada/administração & dosagem , Aminoácidos de Cadeia Ramificada/farmacologia , Animais , Carcinogênese/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ratos , Ratos Endogâmicos ACI
11.
Nat Metab ; 2(8): 657-658, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32694826
12.
Cell Stem Cell ; 16(6): 627-38, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-26046762

RESUMO

Epithelium of the colon and intestine are renewed every 3 days. In the intestine there are at least two principal stem cell pools. The first contains rapid cycling crypt-based columnar (CBC) Lgr5(+) cells, and the second is composed of slower cycling Bmi1-expressing cells at the +4 position above the crypt base. In the colon, however, the identification of Lgr5(-) stem cell pools has proven more challenging. Here, we demonstrate that the intermediate filament keratin-19 (Krt19) marks long-lived, radiation-resistant cells above the crypt base that generate Lgr5(+) CBCs in the colon and intestine. In colorectal cancer models, Krt19(+) cancer-initiating cells are also radioresistant, while Lgr5(+) stem cells are radiosensitive. Moreover, Lgr5(+) stem cells are dispensable in both the normal and neoplastic colonic epithelium, as ablation of Lgr5(+) stem cells results in their regeneration from Krt19-expressing cells. Thus, Krt19(+) stem cells are a discrete target relevant for cancer therapy.


Assuntos
Colo/patologia , Intestinos/patologia , Queratina-19/metabolismo , Células-Tronco Neoplásicas/metabolismo , Tolerância a Radiação , Receptores Acoplados a Proteínas G/metabolismo , Animais , Queratina-19/genética , Camundongos Transgênicos , Células-Tronco Neoplásicas/patologia , Transporte de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
Cancer Res ; 70(21): 8435-45, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20959488

RESUMO

Chronic infectious diseases, such as Helicobacter pylori infection, can promote cancer in a large part through induction of chronic inflammation. Oncogenic K-ras mutation in epithelial cells activates inflammatory pathways, which could compensate for a lack of infectious stimulus. Gastric histopathology and putative progenitor markers [doublecortin and calcium/calmodulin-dependent protein kinase-like 1 (Dcamkl1) and keratin 19 (K19)] in K19-K-ras-V12 (K19-kras) transgenic mice were assessed at 3, 6, 12, and 18 months of age, in comparison with Helicobacter felis-infected wild-type littermates. Inflammation was evaluated by reverse transcription-PCR of proinflammatory cytokines, and K19-kras mice were transplanted with green fluorescent protein (GFP)-labeled bone marrow. Both H. felis infection and K-ras mutation induced upregulation of proinflammatory cytokines, expansion of Dcamkl1(+) cells, and progression to oxyntic atrophy, metaplasia, hyperplasia, and high-grade dysplasia. K19-kras transgenic mice uniquely displayed mucous metaplasia as early as 3 months and progressed to high-grade dysplasia and invasive intramucosal carcinoma by 20 months. In bone marrow-transplanted K19-kras mice that progressed to dysplasia, a large proportion of stromal cells were GFP(+) and bone marrow-derived, but only rare GFP(+) epithelial cells were observed. GFP(+) bone marrow-derived cells included leukocytes and CD45(-) stromal cells that expressed vimentin or α smooth muscle actin and were often found surrounding clusters of Dcamkl1(+) cells at the base of gastric glands. In conclusion, the expression of mutant K-ras in K19(+) gastric epithelial cells can induce chronic inflammation and promote the development of dysplasia.


Assuntos
Carcinoma in Situ/etiologia , Inflamação/etiologia , Mutação/genética , Lesões Pré-Cancerosas/etiologia , Proteínas Proto-Oncogênicas/fisiologia , Células-Tronco/patologia , Neoplasias Gástricas/etiologia , Proteínas ras/fisiologia , Actinas/metabolismo , Animais , Western Blotting , Transplante de Medula Óssea , Carcinoma in Situ/patologia , Proliferação de Células , Quimiocinas/metabolismo , Citocinas/metabolismo , Citometria de Fluxo , Imunofluorescência , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Infecções por Helicobacter/virologia , Helicobacter felis , Humanos , Hiperplasia/etiologia , Hiperplasia/patologia , Hiperplasia/virologia , Técnicas Imunoenzimáticas , Hibridização In Situ , Inflamação/patologia , Queratina-19/genética , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Metaplasia/etiologia , Metaplasia/patologia , Metaplasia/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso/citologia , Músculo Liso/metabolismo , Invasividade Neoplásica , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas p21(ras) , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Células-Tronco/virologia , Estômago/patologia , Estômago/virologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/virologia , Células Estromais/metabolismo , Células Estromais/patologia , Células Estromais/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA