Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G696-704, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26939869

RESUMO

Stromal cells influence epithelial function in both health and disease. Myofibroblasts are abundant stromal cells that influence the cellular microenvironment by release of extracellular matrix (ECM) proteins, growth factors, proteases, cytokines, and chemokines. Cancer-associated myofibroblasts (CAMs) differ from adjacent tissue (ATMs) and normal tissue myofibroblasts (NTMs), but the basis of this is incompletely understood. We report now the differential expression of miRNAs in gastric cancer CAMs. MicroRNA arrays identified differences in the miRNA profile in gastric and esophageal NTMs and in CAMs from stomach compared with NTMs. miR-181d was upregulated in gastric CAMs. Analysis of differentially regulated miRNAs indicated an involvement in Wnt signaling. Examination of a microarray data set then identified Wnt5a as the only consistently upregulated Wnt ligand in gastric CAMs. Wnt5a stimulated miR-181d expression, and knockdown of miR-181d inhibited Wnt5a stimulation of CAM proliferation and migration. Analysis of miR-181d targets suggested a role in chemotaxis. Conditioned medium from CAMs stimulated gastric cancer cell (AGS) migration more than that from ATMs, and miR-181d knockdown reduced the effect of CAM-CM on AGS cell migration but had no effect on AGS cell responses to ATM conditioned media. The data suggest that dysregulation of miRNA expression in gastric CAMs, secondary to Wnt5a signaling, accounts at least in part for the effect of CAMs in promoting cancer cell migration.


Assuntos
MicroRNAs/genética , Miofibroblastos/metabolismo , Neoplasias Gástricas/metabolismo , Via de Sinalização Wnt , Proliferação de Células , Células Cultivadas , Quimiotaxia , Humanos , Miofibroblastos/fisiologia , Neoplasias Gástricas/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
2.
Am J Physiol Gastrointest Liver Physiol ; 309(2): G78-86, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25977510

RESUMO

The pyloric antral hormone gastrin plays a role in remodeling of the gastric epithelium, but the specific targets of gastrin that mediate these effects are poorly understood. Glandular epithelial cells of the gastric corpus express matrix metalloproteinase (MMP)-1, which is a potential determinant of tissue remodeling; some of these cells express the CCK-2 receptor at which gastrin acts. We have now examined the hypothesis that gastrin stimulates expression of MMP-1 in the stomach. We determined MMP-1 transcript abundance in gastric mucosal biopsies from Helicobacter pylori negative human subjects with normal gastric mucosal histology, who had a range of serum gastrin concentrations due in part to treatment with proton pump inhibitors (PPI). The effects of gastrin were studied on gastric epithelial AGS-GR cells using Western blot and migration assays. In human subjects with increased serum gastrin due to PPI usage, MMP-1 transcript abundance was increased 2-fold; there was also increased MMP-7 transcript abundance but not MMP-3. In Western blots, gastrin increased proMMP-1 abundance, as well that of a minor band corresponding to active MMP-1, in the media of AGS-GR cells, and the response was mediated by protein kinase C and p42/44 MAP kinase. There was also increased MMP-1 enzyme activity. Gastrin-stimulated AGS-GR cell migration in both scratch wound and Boyden chamber assays was inhibited by MMP-1 immunoneutralization. We conclude that MMP-1 expression is a target of gastrin implicated in mucosal remodeling.


Assuntos
Movimento Celular , Células Epiteliais/enzimologia , Mucosa Gástrica/enzimologia , Gastrinas/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Animais , Estudos de Casos e Controles , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Mucosa Gástrica/efeitos dos fármacos , Gastrinas/sangue , Gastrinas/genética , Humanos , Metaloproteinase 1 da Matriz/genética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Inibidores da Bomba de Prótons/farmacologia , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais , Transfecção , Regulação para Cima
3.
Carcinogenesis ; 35(8): 1798-806, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24710625

RESUMO

Stromal cells influence cancer progression. Myofibroblasts are an important stromal cell type, which influence the tumour microenvironment by release of extracellular matrix (ECM) proteins, proteases, cytokines and chemokines. The mechanisms of secretion are poorly understood. Here, we describe the secretion of marker proteins in gastric cancer and control myofibroblasts in response to insulin-like growth factor (IGF) stimulation and, using functional genomic approaches, we identify proteins influencing the secretory response. IGF rapidly increased myofibroblast secretion of an ECM protein, TGFßig-h3. The secretory response was not blocked by inhibition of protein synthesis and was partially mediated by increased intracellular calcium (Ca(2+)). The capacity for evoked secretion was associated with the presence of dense-core secretory vesicles and was lost in cells from patients with advanced gastric cancer. In cells responding to IGF-II, the expression of neuroendocrine marker proteins, including secretogranin-II and proenkephalin, was identified by gene array and LC-MS/MS respectively, and verified experimentally. The expression of proenkephalin was decreased in cancers from patients with advanced disease. Inhibition of secretogranin-II expression decreased the secretory response to IGF, and its over-expression recovered the secretory response consistent with a role in secretory vesicle biogenesis. We conclude that normal and some gastric cancer myofibroblasts have a neuroendocrine-like phenotype characterized by Ca(2+)-dependent regulated secretion, dense-core secretory vesicles and expression of neuroendocrine marker proteins; loss of the phenotype is associated with advanced cancer. A failure to regulate myofibroblast protein secretion may contribute to cancer progression.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Miofibroblastos/patologia , Sistemas Neurossecretores/patologia , Secretogranina II/metabolismo , Neoplasias Gástricas/patologia , Western Blotting , Estudos de Casos e Controles , Células Cultivadas , Progressão da Doença , Exocitose/fisiologia , Mucosa Gástrica/metabolismo , Humanos , Técnicas Imunoenzimáticas , Marcação por Isótopo , Miofibroblastos/metabolismo , Sistemas Neurossecretores/metabolismo , Fenótipo , RNA Interferente Pequeno/genética , Secretogranina II/antagonistas & inibidores , Secretogranina II/genética , Neoplasias Gástricas/metabolismo , Espectrometria de Massas em Tandem
4.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G814-22, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23494120

RESUMO

Gastric mucosal health is maintained in response to potentially damaging luminal factors. Aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs) disrupt protective mechanisms leading to bleeding and ulceration. The plasminogen activator system has been implicated in fibrinolysis following gastric ulceration, and an inhibitor of this system, plasminogen activator inhibitor (PAI)-1, is expressed in gastric epithelial cells. In Helicobacter pylori-negative patients with normal gastric histology taking aspirin or NSAIDs, we found elevated gastric PAI-1 mRNA abundance compared with controls; the increase in patients on aspirin was independent of whether they were also taking proton pump inhibitors. In the same patients, aspirin tended to lower urokinase plasminogen activator mRNA. Immunohistochemistry indicated PAI-1 localization to epithelial cells. In a model system using MKN45 or AGS-GR cells transfected with a PAI-1 promoter-luciferase reporter construct, we found no evidence for upregulation of PAI-1 expression by indomethacin, and, in fact, cyclooxygenase products such as PGE2 and PGI2 weakly stimulated expression. Increased gastric PAI-1 mRNA was also found in mice following gavage with ethanol or indomethacin, but plasma PAI-1 was unaffected. In PAI-1(-/-) mice, gastric hemorrhagic lesions in response to ethanol or indomethacin were increased compared with C57BL/6 mice. In contrast, in PAI-1-H/Kß mice in which PAI-1 is overexpressed in parietal cells, there were decreased lesions in response to ethanol and indomethacin. Thus, PAI-1 expression is increased in gastric epithelial cells in response to mucosal irritants such as aspirin and NSAIDs probably via an indirect mechanism, and PAI-1 acts as a local autoregulator to minimize mucosal damage.


Assuntos
Mucosa Gástrica/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Aspirina/farmacologia , Dinoprostona , Etanol/toxicidade , Feminino , Humanos , Indometacina/toxicidade , Masculino , Camundongos , Inibidor 1 de Ativador de Plasminogênio/biossíntese , RNA Mensageiro/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
5.
Carcinogenesis ; 33(8): 1553-62, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22610072

RESUMO

Tumor progression has been linked to changes in the stromal environment. Myofibroblasts are stromal cells that are often increased in tumors but their contribution to cancer progression is not well understood. Here, we show that the secretomes of myofibroblasts derived from gastric cancers [cancer-associated myofibroblasts (CAMs)] differ in a functionally significant manner from those derived from adjacent tissue [adjacent tissue myofibroblasts (ATMs)]. CAMs showed increased rates of migration and proliferation compared with ATMs or normal tissue myofibroblasts (NTMs). Moreover, conditioned medium (CM) from CAMs significantly stimulated migration, invasion and proliferation of gastric cancer cells compared with CM from ATMs or NTMs. Proteomic analysis of myofibroblast secretomes revealed decreased abundance of the extracellular matrix (ECM) adaptor protein like transforming growth factor-ß-induced gene-h3 (TGFßig-h3) in CAMs, which was correlated with lymph node involvement and shorter survival. TGFßig-h3 inhibited IGF-II-stimulated migration and proliferation of both cancer cells and myofibroblasts, and suppressed IGF-II activation of p42/44 MAPkinase; TGFßig-h3 knockdown increased IGF-II- and CM-stimulated migration. Furthermore, administration of TGFßig-h3 inhibited myofibroblast-stimulated growth of gastric cancer xenografts. We conclude that stromal cells exert inhibitory as well as stimulatory effects on tumor cells; TGFßig-h3 is a stromal inhibitory factor that is decreased with progression of gastric cancers.


Assuntos
Mucosa Gástrica/metabolismo , Miofibroblastos/metabolismo , Fatores de Crescimento Transformadores/metabolismo , Progressão da Doença , Humanos , Imuno-Histoquímica , Estômago/patologia
6.
Pflugers Arch ; 463(3): 459-75, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22138972

RESUMO

Myofibroblasts play central roles in wound healing, deposition of the extracellular matrix and epithelial function. Their functions depend on migration and proliferation within the subepithelial matrix, which results in accelerated cellular metabolism. Upregulated metabolic pathways generate protons which need to be excreted to maintain intracellular pH (pH(i)). We isolated human gastric myofibroblasts (HGMs) from surgical specimens of five patients. Then we characterized, for the first time, the expression and functional activities of the Na(+)/H(+) exchanger (NHE) isoforms 1, 2 and 3, and the functional activities of the Na(+)/HCO(3)(-) cotransporter (NBC) and the anion exchanger (AE) in cultured HGMs using microfluorimetry, immunocytochemistry, reverse transcription polymerase chain reaction and immunoblot analysis. We showed that NHE1-3, NBC and AE activities are present in HGMs and that NHE1 is the most active of the NHEs. In scratch wound assays we also demonstrated (using the selective NHE inhibitor HOE-642) that carbachol and insulin like growth factor II (IGF-II) partly stimulate migration of HGMs in a NHE1-dependent manner. EdU incorporation assays revealed that IGF-II induces proliferation of HGMs which is inhibited by HOE-642. The results indicate that NHE1 is necessary for IGF-II-induced proliferation response of HGMs. Overall, we have characterized the pH(i) regulatory mechanisms of HGMs. In addition, we demonstrated that NHE1 activity contributes to both IGF-II- and carbachol-stimulated migration and that it is obligatory for IGF-II-induced proliferation of HGMs.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Miofibroblastos/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Adulto , Idoso , Antiporters/biossíntese , Carbacol/farmacologia , Proteínas de Transporte de Cátions/antagonistas & inibidores , Movimento Celular , Proliferação de Células , Feminino , Guanidinas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Fator de Crescimento Insulin-Like II/fisiologia , Masculino , Simportadores de Sódio-Bicarbonato/fisiologia , Trocador 1 de Sódio-Hidrogênio , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/biossíntese , Estômago/citologia , Sulfonas/farmacologia
7.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G446-53, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21193525

RESUMO

Plasminogen activator inhibitor (PAI)-1 is associated with cancer progression, fibrosis and thrombosis. It is expressed in the stomach but the mechanisms controlling its expression there, and its biological role, are uncertain. We sought to define the role of gastrin in regulating PAI-1 expression and to determine the relevance for gastrin-stimulated cell migration and invasion. In gastric biopsies from subjects with elevated plasma gastrin, the abundances of PAI-1, urokinase plasminogen activator (uPA), and uPA receptor (uPAR) mRNAs measured by quantitative PCR were increased compared with subjects with plasma concentrations in the reference range. In patients with hypergastrinemia due to autoimmune chronic atrophic gastritis, there was increased abundance of PAI-1, uPA, and uPAR mRNAs that was reduced by octreotide or antrectomy. Immunohistochemistry revealed localization of PAI-1 to parietal cells and enterochromaffin-like cells in micronodular neuroendocrine tumors in hypergastrinemic subjects. Transcriptional mechanisms were studied by using a PAI-1-luciferase promoter-reporter construct transfected into AGS-G(R) cells. There was time- and concentration-dependent increase of PAI-1-luciferase expression in response to gastrin that was reversed by inhibitors of the PKC and MAPK pathways. In Boyden chamber assays, recombinant PAI-1 inhibited gastrin-stimulated AGS-G(R) cell migration and invasion, and small interfering RNA treatment increased responses to gastrin. We conclude that elevated plasma gastrin concentrations are associated with increased expression of gastric PAI-1, which may act to restrain gastrin-stimulated cell migration and invasion.


Assuntos
Células Epiteliais/metabolismo , Gastrinas/farmacologia , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Celulas Tipo Enterocromafim/metabolismo , Gastrinas/sangue , Humanos , Octreotida , RNA Mensageiro/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Estômago/citologia , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
8.
Gastroenterology ; 138(4): 1514-24, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20080096

RESUMO

BACKGROUND & AIMS: In human beings, as in most vertebrates, the release of the intestinal peptide cholecystokinin (CCK) by ingested food plays a major role both in digestion and the regulation of further food intake, but the changes in brain function and their underlying activation mechanisms remain unknown. Our aim was to explore, using a novel physiologic magnetic resonance imaging approach, the temporospatial brain activation matrix, in response to ingestion of a lipid meal and, by use of a CCK-1 receptor antagonist, to define the role of CCK in this activation. METHODS: We studied, in 19 healthy subjects, the brain activation responses to ingested lipid (dodecanoic acid) or saline (control) with magnetic resonance imaging. Gallbladder volume, plasma CCK levels, and subjective hunger and fullness scores were also recorded. The experiment was then repeated, with and without prior administration of the CCK-1 receptor antagonist dexloxiglumide (600 mg orally) with a controlled, randomized order, latin-square design. RESULTS: Ingested lipid activated bilaterally a matrix of brain areas, particularly the brain stem, pons, hypothalamus, and also cerebellum and motor cortical areas. These activations were abolished by dexloxiglumide, indicating a CCK-mediated pathway, independent of any nutrient-associated awareness cues. CONCLUSION: The identification of these activations now defines the lipid-activated brain matrix and provides a means by which the gut-derived homeostatic mechanisms of food regulation can be distinguished from secondary sensory and hedonic cues, thereby providing a new approach to exploring aberrant human gastrointestinal responses and eating behavior.


Assuntos
Encéfalo/metabolismo , Colecistocinina/fisiologia , Lipídeos/administração & dosagem , Adulto , Índice de Massa Corporal , Colecistocinina/sangue , Feminino , Vesícula Biliar/fisiologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Oxigênio/sangue , Ácidos Pentanoicos/farmacologia
9.
PLoS One ; 9(7): e104877, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25127029

RESUMO

Stromal cells such as myofibroblasts influence tumor progression. The mechanisms are unclear but may involve effects on both tumor cells and recruitment of bone marrow-derived mesenchymal stromal cells (MSCs) which then colonize tumors. Using iTRAQ and LC-MS/MS we identified the adipokine, chemerin, as overexpressed in esophageal squamous cancer associated myofibroblasts (CAMs) compared with adjacent tissue myofibroblasts (ATMs). The chemerin receptor, ChemR23, is expressed by MSCs. Conditioned media (CM) from CAMs significantly increased MSC cell migration compared to ATM-CM; the action of CAM-CM was significantly reduced by chemerin-neutralising antibody, pretreatment of CAMs with chemerin siRNA, pretreatment of MSCs with ChemR23 siRNA, and by a ChemR23 receptor antagonist, CCX832. Stimulation of MSCs by chemerin increased phosphorylation of p42/44, p38 and JNK-II kinases and inhibitors of these kinases and PKC reversed chemerin-stimulated MSC migration. Chemerin stimulation of MSCs also induced expression and secretion of macrophage inhibitory factor (MIF) that tended to restrict migratory responses to low concentrations of chemerin but not higher concentrations. In a xenograft model consisting of OE21 esophageal cancer cells and CAMs, homing of MSCs administered i.v. was inhibited by CCX832. Thus, chemerin secreted from esophageal cancer myofibroblasts is a potential chemoattractant for MSCs and its inhibition may delay tumor progression.


Assuntos
Quimiocinas/metabolismo , Neoplasias Esofágicas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/fisiologia , Miofibroblastos/metabolismo , Animais , Linhagem Celular Tumoral , Quimiotaxia , Neoplasias Esofágicas/patologia , Humanos , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transplante de Neoplasias , Proteína Quinase C/metabolismo , Receptores de Quimiocinas/metabolismo , Migração Transendotelial e Transepitelial
10.
PLoS One ; 8(10): e76462, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098507

RESUMO

INTRODUCTION: Autoimmune chronic atrophic gastritis (CAG) causes hypochlorhydria and hypergastrinaemia, which can lead to enterochromaffin-like (ECL) cell hyperplasia and gastric neuroendocrine tumours (type 1 gastric NETs). Most behave indolently, but some larger tumours metastasise. Antrectomy, which removes the source of the hypergastrinaemia, usually causes tumour regression. Non-clinical and healthy-subject studies have shown that netazepide (YF476) is a potent, highly selective and orally-active gastrin/CCK-2 receptor antagonist. Also, it is effective in animal models of ECL-cell tumours induced by hypergastrinaemia. AIM: To assess the effect of netazepide on tumour biomarkers, number and size in patients with type I gastric NETs. METHODS: We studied 8 patients with multiple tumours and raised circulating gastrin and chromogranin A (CgA) concentrations in an open trial of oral netazepide for 12 weeks, with follow-up 12 weeks later. At 0, 6, 12 and 24 weeks, we carried out gastroscopy, counted and measured tumours, and took biopsies to assess abundances of several ECL-cell constituents. At 0, 3, 6, 9, 12 and 24 weeks, we measured circulating gastrin and CgA and assessed safety and tolerability. RESULTS: Netazepide was safe and well tolerated. Abundances of CgA (p<0.05), histidine decarboxylase (p<0.05) and matrix metalloproteinase-7(p<0.10) were reduced at 6 and 12 weeks, but were raised again at follow-up. Likewise, plasma CgA was reduced at 3 weeks (p<0.01), remained so until 12 weeks, but was raised again at follow-up. Tumours were fewer and the size of the largest one was smaller (p<0.05) at 12 weeks, and remained so at follow-up. Serum gastrin was unaffected. CONCLUSION: The reduction in abundances, plasma CgA, and tumour number and size by netazepide show that type 1 NETs are gastrin-dependent tumours. Failure of netazepide to increase serum gastrin further is consistent with achlorhydria. Netazepide is a potential new treatment for type 1 NETs. Longer, controlled trials are justified. TRIAL REGISTRATION: European Union EudraCT database 2007-002916-24 https://www.clinicaltrialsregister.eu/ctr-search/search?query=2007-002916-24ClinicalTrials.gov NCT01339169 http://clinicaltrials.gov/ct2/show/NCT01339169?term=yf476&rank=5.


Assuntos
Benzodiazepinonas/uso terapêutico , Gastrite Atrófica/complicações , Tumores Neuroendócrinos/complicações , Tumores Neuroendócrinos/tratamento farmacológico , Compostos de Fenilureia/uso terapêutico , Receptor de Colecistocinina B/antagonistas & inibidores , Neoplasias Gástricas/complicações , Neoplasias Gástricas/tratamento farmacológico , Idoso , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Benzodiazepinonas/farmacologia , Biomarcadores/sangue , Biomarcadores/metabolismo , Biópsia , Cromogranina A/sangue , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrinas/sangue , Gastroscopia , Humanos , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/patologia , Compostos de Fenilureia/farmacologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Resultado do Tratamento , Carga Tumoral
11.
Eur J Gastroenterol Hepatol ; 25(4): 474-81, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23249603

RESUMO

BACKGROUND: Type-1 gastric neuroendocrine tumours (NETs) arise in some patients with chronic hypergastrinaemia secondary to autoimmune atrophic gastritis. Patients with small tumours are usually managed conservatively, because their prognosis is very good. However, larger tumours may require surgical intervention. Many type-1 gastric NETs regress following antrectomy because this removes the source of hypergastrinaemia. However, some tumours do not regress following antrectomy and additional surgery may be required. An octreotide suppression test has been previously suggested as a means to assess whether type-1 gastric NETs are likely to regress following antrectomy. AIM: To prospectively examine the role of a short-term intravenous octreotide suppression test in predicting type-1 gastric NET regression in five patients who subsequently underwent antrectomy. MATERIALS AND METHODS: Serum gastrin concentrations and gastric corpus and tumour histidine decarboxylase mRNA abundances were assessed in patients with type-1 gastric NETs before and 72 h after the administration of 25 µg/h intravenous octreotide. Gastric tumour response was assessed endoscopically following subsequent antrectomy. RESULTS: All patients showed significant decreases in serum gastrin concentrations as well as corpus and tumour biopsy histidine decarboxylase mRNA abundance following octreotide infusion. All patients also showed resolution of hypergastrinaemia following subsequent antrectomy. However, tumour regression was only observed in four of the five patients. One patient had a persistent tumour 3 years after antrectomy and required additional surgical resection. CONCLUSION: A positive octreotide suppression test result does not always predict response to antrectomy in patients with type-1 gastric NETs. Assessment of gastric mucosal responses to a gastrin/CCK-2 receptor antagonist may therefore also be helpful.


Assuntos
Antineoplásicos Hormonais , Tumores Neuroendócrinos/diagnóstico , Octreotida , Antro Pilórico/cirurgia , Neoplasias Gástricas/diagnóstico , Adulto , Idoso , Antineoplásicos Hormonais/administração & dosagem , Feminino , Gastrectomia/métodos , Gastrinas/sangue , Gastroscopia/métodos , Histidina Descarboxilase/biossíntese , Histidina Descarboxilase/genética , Humanos , Infusões Intravenosas , Masculino , Tumores Neuroendócrinos/cirurgia , Octreotida/administração & dosagem , Seleção de Pacientes , Prognóstico , Estudos Prospectivos , RNA Mensageiro/genética , Neoplasias Gástricas/cirurgia , Resultado do Tratamento
12.
Cancer Res ; 68(23): 9900-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19047171

RESUMO

Global hypomethylation has long been recognized as a feature of the malignant epithelial component in human carcinomas. Here we show evidence for this same type of epigenetic alteration in cancer-associated stromal myofibroblasts. We used methylation-sensitive SNP array analysis (MSNP) to profile DNA methylation in early-passage cultures of stromal myofibroblasts isolated from human gastric cancers. The MSNP data indicated widespread hypomethylation in these cells, with rare focal gains of methylation, conclusions that were independently validated by bisulfite sequencing and by a methylation-sensitive cytosine incorporation assay. Immunohistochemistry with anti-5-methylcytosine (anti-5-methyl-C) in a series of gastrectomy specimens showed frequent loss of methylation in nuclei of both the malignant epithelial cells and alpha-smooth muscle actin (ASMA)-positive stromal myofibroblasts of both intestinal-type and diffuse carcinomas. We confirmed this phenomenon and established its onset at the stage of noninvasive dysplastic lesions by immunohistochemistry for anti-5-methyl-C in a transgenic mouse model of multistage gastric carcinogenesis. These findings indicate similar general classes of epigenetic alterations in carcinoma cells and their accompanying reactive stromal cells and add to accumulating evidence for biological differences between normal and cancer-associated myofibroblasts.


Assuntos
Metilação de DNA , DNA de Neoplasias/genética , Neoplasias Gástricas/genética , Animais , Instabilidade Cromossômica , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/genética , DNA de Neoplasias/metabolismo , Nucleotídeos de Desoxicitosina/genética , Nucleotídeos de Desoxicitosina/metabolismo , Modelos Animais de Doenças , Fibroblastos/patologia , Humanos , Imuno-Histoquímica , Camundongos , Polimorfismo de Nucleotídeo Único , Neoplasias Gástricas/patologia , Células Estromais/patologia
13.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G431-41, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18599586

RESUMO

The gastric pathogen Helicobacter pylori (H. pylori) is linked to peptic ulcer and gastric cancer, but the relevant pathophysiological mechanisms are unclear. We now report that H. pylori stimulates the expression of plasminogen activator inhibitor (PAI)-1, urokinase plasminogen activator (uPA), and its receptor (uPAR) in gastric epithelial cells and the consequences for epithelial cell proliferation. Real-time PCR of biopsies from gastric corpus, but not antrum, showed significantly increased PAI-1, uPA, and uPAR in H. pylori-positive patients. Transfection of primary human gastric epithelial cells with uPA, PAI-1, or uPAR promoters in luciferase reporter constructs revealed expression of all three in H+/K+ATPase- and vesicular monoamine transporter 2-expressing cells; uPA was also expressed in pepsinogen- and uPAR-containing trefoil peptide-1-expressing cells. In each case expression was increased in response to H. pylori and for uPA, but not PAI-1 or uPAR, required the virulence factor CagE. H. pylori also stimulated soluble and cell surface-bound uPA activity, and both were further increased by PAI-1 knockdown, consistent with PAI-1 inhibition of endogenous uPA. H. pylori stimulated epithelial cell proliferation, which was inhibited by uPA immunoneutralization and uPAR knockdown; exogenous uPA also stimulated proliferation that was further increased after PAI-1 knockdown. The proliferative effects of uPA were inhibited by immunoneutralization of the EGF receptor and of heparin-binding EGF (HB-EGF) by the mutant diphtheria toxin CRM197 and an EGF receptor tyrosine kinase inhibitor. H. pylori induction of uPA therefore leads to epithelial proliferation through activation of HB-EGF and is normally inhibited by concomitant induction of PAI-1; treatments directed at inhibition of uPA may slow the progression to gastric cancer.


Assuntos
Proliferação de Células , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/isolamento & purificação , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas de Bactérias/metabolismo , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Feminino , Mucosa Gástrica/enzimologia , Mucosa Gástrica/patologia , Genes Reporter , Infecções por Helicobacter/enzimologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Metaloproteases/metabolismo , Pessoa de Meia-Idade , Inibidor 1 de Ativador de Plasminogênio/genética , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/microbiologia , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Transfecção , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/genética
14.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G347-54, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17463184

RESUMO

The gastric pathogen Helicobacter pylori accelerates the progression to gastric cancer but the precise mechanisms that mediate carcinogenesis remain unidentified. We now describe how Helicobacter and gastrin stimulate the expression of a putative growth factor, Reg1, in primary gastric epithelial cells. RT-PCR and Western immunoblotting of human gastric corpus and antrum showed significantly increased Reg1alpha in H. pylori-infected patients. Similarly, Reg1 was increased in the stomachs of H. felis-infected INS-GAS mice. To study transcriptional regulation of the Reg1 gene, we transfected primary mouse gastric glands with -2111 bp and -104 bp Reg1 promoter-luciferase reporter constructs. Expression of both constructs was detected in pepsinogen- and VMAT-2-expressing cells, which corresponds to the normal pattern of expression of human and mouse endogenous Reg1. The expression of both constructs was increased in response to gastrin and H. pylori, and there were potentiating interactions between them; in contrast, only the -2111 bp construct responded to H. felis. Mutation of a C-rich putative regulatory element within the -104 bp sequence abolished the response to gastrin but not to H. pylori whereas mutation of the proximal -98 to -93 region of the promoter reduced the response to H. pylori but not to gastrin. Stimulation of Reg1 by H. pylori required the virulence factor CagA. These data indicate that expression of the putative growth factor Reg1 is controlled through separate promoter elements by gastrin and Helicobacter.


Assuntos
Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Gastrinas/fisiologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Litostatina/biossíntese , Regiões Promotoras Genéticas/fisiologia , Animais , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Litostatina/genética , Masculino , Camundongos , Camundongos Transgênicos , Antro Pilórico/metabolismo , Ratos
15.
Growth Factors ; 24(1): 45-53, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16393693

RESUMO

Epithelial ovarian cancer (EOC) is the most common and lethal form of gynecological malignancy. These cancers are thought to be derived from the ovarian surface epithelium (OSE). We have previously reported that the epithelial-specific FGF receptor 2 splice variant IIIb is not expressed in normal OSE, but is expressed in approximately 80% of EOCs. We have examined the phenotypic effects of ligands to FGF receptor 2-IIIb, namely FGFs 1,7 and 10, on a panel of EOC cell lines. We show that these ligands increase cell viability, induce DNA synthesis, motility and chemotaxis and protect from spontaneous cell death when EOC cells are maintained in serum free medium. A blocking antiserum to FGF-7 reduces viability of 41-M EOC cells, and abrogates the ability of ascitic fluid containing FGF-7 to induce DNA synthesis in these cells. Finally, we show that FGF-7 can induce a reorganization of the actin cytoskeleton in SK-OV-3 ovarian cancer cells. It is suggested that ligands to FGF receptor 2-IIIb affect a range of phenotypes important in the neoplastic growth of EOCs.


Assuntos
Citoesqueleto/fisiologia , Células Epiteliais/patologia , Neoplasias Ovarianas/patologia , Ovário/patologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Actinas/fisiologia , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Quimiotaxia , Meios de Cultura Livres de Soro , Replicação do DNA , Células Epiteliais/metabolismo , Feminino , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 7 de Crescimento de Fibroblastos/metabolismo , Humanos , Ligantes , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo
16.
Gastroenterology ; 130(6): 1754-63, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16697739

RESUMO

BACKGROUND & AIMS: Interactions between epithelial and stromal cells are important determinants of mucosal organization, but the signaling mechanisms are understood incompletely. Matrix metalloproteinase (MMP)-7 is produced uniquely in epithelia, may act on growth factors and matrix proteins, and in the stomach is increased with Helicobacter pylori infection. We have studied the role of MMP-7 in signaling between epithelial cells and a key stromal cell type, the myofibroblast. METHODS: Immunohistochemistry and Western blotting were applied to gastric corpus biopsy specimens; primary cultures of human gastric glands and myofibroblasts were used to study the role of MMP-7 in regulating proliferation and migration of the latter, and MMP-7 substrates were identified by proteomic methods. RESULTS: Increased abundance of the myofibroblast marker alpha-smooth muscle actin was identified in H. pylori-positive biopsy specimens. Media from H pylori-infected gastric epithelial cultures stimulated proliferation and migration of primary human gastric myofibroblasts and antisense oligonucleotide treatment indicated a role for MMP-7. Proteomic methods identified insulin-like growth factor binding protein (IGFBP)-5 as a substrate for MMP-7 in medium from gastric myofibroblasts. Knockdown of IGFBP-5 by small interfering RNA or immunoneutralization of IGF-II, abolished myofibroblast responses to MMP-7. Proliferation of gastric epithelial cells also was stimulated by MMP-7-treated myofibroblasts via IGF-II. CONCLUSIONS: MMP-7 acts as an epithelial-derived signal increasing the bioavailability of IGF-II released from myofibroblasts. Because IGF-II acts on both stromal and epithelial cells, the findings suggest that increased MMP-7 expression contributes to redefining the niche occupied by dividing cells and leading to hyperproliferation in H pylori infection.


Assuntos
Fibroblastos/citologia , Mucosa Gástrica/microbiologia , Gastrinas/análise , Infecções por Helicobacter/patologia , Helicobacter pylori/citologia , Metaloproteinase 7 da Matriz/metabolismo , Animais , Biópsia por Agulha , Western Blotting , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Mucosa Gástrica/patologia , Gastrinas/biossíntese , Infecções por Helicobacter/fisiopatologia , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 7 da Matriz/análise , Camundongos , Probabilidade , Radioimunoensaio , Sensibilidade e Especificidade
17.
Exp Cell Res ; 288(2): 390-402, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12915130

RESUMO

Epithelial ovarian cancer is the most common form of gynaecological malignancy. This lethal disease is thought to arise in ovarian surface epithelial (OSE) cells. The biology of these cells is not well understood, due to the limited amount of tissue that can be obtained from a single biopsy and their limited life span in culture. To overcome these problems, we have conditionally immortalised OSE cells with the catalytic subunit of telomerase (hTERT) and a temperature-sensitive form of SV40 Large T antigen (tsT). We have maintained these cells (designated OSE-C2) in culture for more than 100 population doublings after introduction of the immortalising genes. Early passage OSE-C2 cells have a near-tetraploid karyotype and exhibit a dual mesenchymal-epithelial phenotype, with consistent expression of vimentin and variable expression of cytokeratins and type III collagen, and absence of E cadherin expression. OSE-C2 cells proliferate steadily at the permissive temperature of 33 degrees C, but fail to increase in number at the nonpermissive temperature of 39 degrees C. Serum-deprived OSE-C2 cells are stimulated to grow at 33 degrees C by EGF, whereas they are growth inhibited at 33 degrees C by TGFbeta in the presence or the absence of serum. When temperature shifted to the nonpermissive temperature, OSE-C2 cells modulate to a more mesenchymal phenotype, and a proportion of the cells undergo senescence and/or apoptosis. Moreover, at the nonpermissive temperature, the levels of p53 and SV40 Large T antigen diminish, whilst the level of p21 increases, whereas the level of p16 and telomerase activity is unchanged. This experimental system shows that expression of telomerase alone only allows limited proliferative potential of OSE cells; expression of tsT is necessary to maintain these cells in culture for longer periods, perhaps by its ability to inactivate components of the p53/Rb pathway. OSE-C2 cells may be useful in studying the physiology and differentiation of human OSE cells and provide insight into the poorly understood earliest stages of epithelial ovarian cancer.


Assuntos
Antígenos Transformantes de Poliomavirus/metabolismo , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Ovário/citologia , Telomerase/metabolismo , Antígenos Transformantes de Poliomavirus/genética , Apoptose/fisiologia , Biomarcadores , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Linhagem Celular Transformada , Células Cultivadas , Senescência Celular/fisiologia , Proteínas de Ligação a DNA , Células Epiteliais/metabolismo , Feminino , Substâncias de Crescimento/metabolismo , Humanos , Cariotipagem , Ovário/metabolismo , Telomerase/genética , Telômero/metabolismo , Temperatura , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA