Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Gastroenterology ; 141(2): 731-41, 741.e1-4, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21703267

RESUMO

BACKGROUND & AIMS: Animal studies have indicated that pancreatic exocrine acinar cells have phenotypic plasticity. In rodents, acinar cells can differentiate into ductal precursors that can be converted to pancreatic ductal adenocarcinoma or insulin-producing endocrine cells. However, little is known about human acinar cell plasticity. We developed nongenetic and genetic lineage tracing methods to study the fate of human acinar cells in culture. METHODS: Human exocrine tissue was obtained from organ donors, dissociated, and cultured. Cell proliferation and survival were measured, and cell phenotypes were analyzed by immunocytochemistry. Nongenetic tracing methods were developed based on selective binding and uptake by acinar cells of a labeled lectin (Ulex europaeus agglutinin 1). Genetic tracing methods were developed based on adenoviral introduction of a Cre-lox reporter system, controlled by the amylase promoter. RESULTS: Both tracing methods showed that human acinar cells can transdifferentiate into cells that express specific ductal markers, such as cytokeratin 19, hepatocyte nuclear factor 1ß, SOX9, CD133, carbonic anhydrase II, and cystic fibrosis transmembrane conductance regulator. Within 1 week of culture, all surviving acinar cells had acquired a ductal phenotype. This transdifferentiation was decreased by inhibiting mitogen-activated protein kinase signaling. CONCLUSIONS: Human acinar cells have plasticity similar to that described in rodent cells. These results might be used to develop therapeutic strategies for patients with diabetes or pancreatic cancer.


Assuntos
Linhagem da Célula/genética , Transdiferenciação Celular/genética , Pâncreas Exócrino/citologia , Ductos Pancreáticos/citologia , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Antígeno AC133 , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Anidrase Carbônica II/metabolismo , Linhagem da Célula/fisiologia , Proliferação de Células , Sobrevivência Celular , Transdiferenciação Celular/fisiologia , Células Cultivadas , Quimotripsina/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Genes Reporter , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Queratina-19/metabolismo , Antígeno Ki-67/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pâncreas Exócrino/metabolismo , Ductos Pancreáticos/metabolismo , Peptídeos/metabolismo , Fenótipo , Lectinas de Plantas/farmacocinética , Regiões Promotoras Genéticas , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/genética , Transdução Genética
2.
Diabetes ; 51(3): 686-90, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11872667

RESUMO

It is still unclear which factors regulate pancreatic regeneration and beta-cell neogenesis and which precursor cells are involved. We evaluated the role of intravenously infused gastrin in regenerating pancreas of duct-ligated rats. The ligation of exocrine ducts draining the splenic half of the pancreas resulted in acinoductal transdifferentiation within the ligated part but not in the unligated part. We found that infusion of gastrin from day 7 to 10 postligation resulted in a doubling of the beta-cell mass in the ligated part as measured by morphometry. This increase in insulin-expressing cells was not associated with increased proliferation, hypertrophy, or reduced cell death of the beta-cells. Furthermore, we found an increased percentage of single, extra-insular beta-cells and small beta-cell clusters induced by gastrin infusion. These changes occurred only in the ligated part of the pancreas, where transdifferentiation of the exocrine acinar cells to ductlike cells (metaplasia) had occurred, and was not found in the normal unaffected pancreatic tissue. In conclusion, we demonstrate that administration of gastrin stimulates beta-cell neogenesis and expansion of the beta-cell mass from transdifferentiated exocrine pancreas.


Assuntos
Gastrinas/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Glicemia/análise , Diferenciação Celular/efeitos dos fármacos , Imuno-Histoquímica , Ilhotas Pancreáticas/anatomia & histologia , Ilhotas Pancreáticas/fisiologia , Ligadura , Masculino , Tamanho do Órgão/efeitos dos fármacos , Pâncreas/anatomia & histologia , Pâncreas/fisiologia , Ductos Pancreáticos/cirurgia , Ratos , Ratos Wistar , Regeneração , Tioléster Hidrolases/análise , Ubiquitina Tiolesterase
3.
Virchows Arch ; 444(1): 61-5, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14648221

RESUMO

Injured pancreatic tissue, for example, after duct ligation, undergoes remodeling, which involves the replacement of exocrine acini by duct-like structures. This acinoductal metaplasia is probably at least partly due to transdifferentiation of amylase-positive, cytokeratin-20 (CK20)-negative acinar cells into amylase-negative, CK20-positive duct-like cells. Due to the kinetics of these phenotypic changes, however, it has not been possible to demonstrate transitional stages of differentiation, which would express both markers at the same time. We took advantage of the fact that dexamethasone treatment inhibits the loss of amylase from acinar cells to demonstrate transitional cells co-expressing amylase and CK20. This was found both in vivo, where duct-ligation induced metaplasia, and in vitro, after isolation of acini. In addition, we found evidence for an acinar-to-islet conversion under the form of transitional cells co-expressing amylase and insulin. These observations strengthen the notion that fully differentiated cells, such as exocrine pancreatic cells, retain the capacity to undergo important phenotypic switches. This finding could have applications in tissue engineering or cell replacement strategies.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Pâncreas/citologia , Amilases/análise , Animais , Células Cultivadas , Fluoresceína-5-Isotiocianato , Imunofluorescência , Corantes Fluorescentes , Imuno-Histoquímica , Insulina/análise , Proteínas de Filamentos Intermediários/análise , Queratina-20 , Queratinas , Masculino , Pâncreas/química , Pâncreas/efeitos dos fármacos , Fenótipo , Ratos , Ratos Wistar , Rodaminas
4.
Pancreas ; 36(1): e1-6, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18192867

RESUMO

OBJECTIVES: Many efforts are spent in identifying stem cells in adult pancreas because these could provide a source of beta cells for cell-based therapy of type 1 diabetes. Prominin-1, particularly its specific glycosylation-dependent AC133 epitope, is expressed on stem/progenitor cells of various human tissues and can be used to isolate them. We, therefore, examined its expression in adult human pancreas. METHODS: To detect prominin-1 protein, monoclonal antibody CD133/1 (AC133 clone), which recognizes the AC133 epitope, and the alphahE2 antiserum, which is directed against the human prominin-1 polypeptide, were used. Prominin-1 RNA expression was analyzed by real-time polymerase chain reaction. RESULTS: We report that all duct-lining cells of the pancreas express prominin-1. Most notably, the cells that react with the alphahE2 antiserum also react with the AC133 antibody. After isolation and culture of human exocrine cells, we found a relative increase in prominin-1 expression both at protein and RNA expression level, which can be explained by an enrichment of cells with ductal phenotype in these cultures. CONCLUSIONS: Our data show that pancreatic duct cells express prominin-1 and surprisingly reveal that its particular AC133 epitope is not an exclusive stem and progenitor cell marker.


Assuntos
Antígenos CD/análise , Biomarcadores/análise , Glicoproteínas/análise , Ductos Pancreáticos/química , Peptídeos/análise , Células-Tronco/química , Antígeno AC133 , Anticorpos Monoclonais , Antígenos CD/genética , Antígenos CD/imunologia , Células Cultivadas , Epitopos/imunologia , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Imuno-Histoquímica , Microscopia Eletrônica , Microscopia de Fluorescência , Peptídeos/genética , Peptídeos/imunologia , Reação em Cadeia da Polimerase , RNA Mensageiro/análise
5.
Am J Pathol ; 169(4): 1206-14, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17003479

RESUMO

When pancreatic tissue is injured after duct obstruction, acinoductal metaplasia is observed. Similar metaplastic changes occur when exocrine pancreatic cells are isolated and cultured. We demonstrate that under these experimental conditions the exocrine acinar cells lose their differentiated characteristics: expression of the acinar transcription factors p48/Ptf1alpha and Mist1 is decreased or lost, whereas expression of the embryonic transcription factor Pdx1 is increased. The receptors Notch1 and Notch2, members of the DSL family of Notch ligands, and the target genes in the Notch-signaling pathway Hes1, Hey1, and Hey2 become strongly up-regulated. We noted also reduced expression of Sel1L, a Notch repressor that is normally highly expressed in exocrine pancreas. Stimulation of Notch by its ligand Jagged1 diminished the proliferation of cultured metaplastic exocrine cells. Chemical inhibition of Notch signaling resulted in increased proliferation and induction of the cell-cycle regulator p21Cip1. This effect seems to be Hes1-independent and mainly coincides with decreased Hey1 and Hey2 mRNA expression. In conclusion, we demonstrate that during acinoductal metaplasia the Notch-signaling pathway is activated concomitantly with changes in transcription factor expression of pancreatic acinar cells. In addition, we show that Notch signaling is implicated in the suppression of proliferation of these metaplastic exocrine cells. The latter may be important in protection from neoplastic transformation.


Assuntos
Pâncreas Exócrino/patologia , Ductos Pancreáticos/patologia , Receptores Notch/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/análise , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/análise , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Homeodomínio/análise , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Ligadura , Masculino , Metaplasia/genética , Metaplasia/metabolismo , Metaplasia/patologia , Pâncreas Exócrino/química , Pâncreas Exócrino/metabolismo , Ductos Pancreáticos/química , Ductos Pancreáticos/metabolismo , Ratos , Ratos Wistar , Receptores Notch/análise , Receptores Notch/genética , Proteínas Repressoras/análise , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transativadores/análise , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição HES-1 , Fatores de Transcrição/análise , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima
6.
Differentiation ; 73(6): 278-86, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16138828

RESUMO

There is currently much interest in the possibility to treat chronic diseases by cell replacement or regenerative therapies. Most of these studies focus on the manipulation of undifferentiated stem cells. However, tissue repair and regeneration can also be achieved by differentiated cells, which, in certain conditions, can even transdifferentiate to other cell types. Such transdifferentiations can lead to tissue metaplasia. The pancreas is an organ wherein metaplasia has been well investigated and for which experimental models have been recently developed allowing to unravel the molecular basis of transdifferentiation. Pancreatic metaplasias studied so far include the conversion of exocrine acinar cells to duct cells, exocrine cells to endocrine islet cells, endocrine cells to duct cells, and acinar cells to hepatocytes. Epitheliomesenchymal transitions have also been described. The available evidence indicates that mature cells can be reprogrammed by specific environmental cues inducing the expression of cell type-specific transcription factors. For example, the glucocorticoid hormone dexamethasone induces pancreatic transdifferentiation to hepatocytes, whereas the combination of epidermal growth factor and leukemia-inhibitory factor induces exocrine-endocrine transdifferentiation in vitro. Further unravelling of the involved signal transduction pathways, transcription factor networks, and chromatin modifications is required to manipulate metaplasia at will and to apply it in tissue repair or regeneration.


Assuntos
Pâncreas/embriologia , Pâncreas/patologia , Pancreatopatias/etiologia , Animais , Diferenciação Celular , Humanos , Metaplasia/embriologia , Metaplasia/etiologia , Pancreatopatias/patologia
7.
Histochem Cell Biol ; 117(6): 535-40, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12107504

RESUMO

Nestin is an intermediate filament protein expressed by neuroepithelial stem cells and which has been proposed to represent also a marker for putative islet stem cells. The aim of this study was to characterize the cell type(s) expressing nestin in the rat pancreas. By immunohistochemistry, nestin positivity was localized exclusively in mesenchymal cells of normal and regenerating adult pancreas. In the latter condition, the number of nestin-positive cells and the intensity of nestin immunoreactivity were greatly increased. Most nestin-positive cells had the morphology of stellate cells, a type of pericyte associated with blood vessels which has been previously reported to occur in liver and pancreas. In addition, nestin positivity was present in endothelial cells from neocapillaries during pancreas regeneration, and in all blood vessels during morphogenesis in fetal pancreas. Nestin expression was not found in the ductal epithelial cells from which islet cells originate in fetal and regenerating pancreas. In primary pancreatic tissue explants, nestin-positive mesenchymal cells rapidly attached to plastic and proliferated. These cells also expressed desmin, vimentin, and glial fibrillary acidic protein which are known to represent stellate cell markers. In summary, nestin in the pancreas is primarily a marker for reactive stellate cells, or pericytes, and endothelial cells during active angiogenesis.


Assuntos
Endotélio Vascular/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Neovascularização Fisiológica , Proteínas do Tecido Nervoso , Pâncreas/citologia , Pericitos/metabolismo , Animais , Capilares/citologia , Capilares/crescimento & desenvolvimento , Embrião de Mamíferos , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Imuno-Histoquímica , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Morfogênese , Nestina , Ratos , Ratos Wistar , Regeneração , Distribuição Tecidual
8.
Hepatology ; 39(6): 1499-507, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15185290

RESUMO

Under certain experimental conditions, hepatocytes can arise in the pancreas. It has been suggested that the pancreas retains a source of hepatocyte progenitor cells. However, such cells have not been yet identified in the adult pancreas. We describe here the transdifferentiation of primary rat pancreatic exocrine cells into hepatocyte-like cells during 5 days of tissue culture in the presence of dexamethasone (DX). Using reverse-transcription polymerase chain reaction and immunocytochemistry, it was observed that DX treatment induced albumin RNA and protein expression in the cells. Coexpression of albumin and amylase, and the absence of cell proliferation, demonstrated a direct transdifferentiation of acinar cells to hepatocytic cells. CCAAT enhancer-binding protein-ss protein, a liver-enriched transcription factor that is considered to be the master switch in pancreatohepatic transdifferentiation, and alpha-fetoprotein were markedly upregulated in the cells after treatment with DX. We compared transcriptional profiles of freshly isolated exocrine cells and DX-treated cells using oligonucleotide microarrays and found that multiple liver-specific genes are induced along with albumin, and that certain pancreatic genes are downregulated in the DX-treated cells. In conclusion, these observations support the notion of plasticity in the adult pancreas and that exocrine cells can be reprogrammed to transdifferentiate into other cell types such as hepatocytes.


Assuntos
Pâncreas/citologia , Fatores Etários , Albuminas/biossíntese , Animais , Diferenciação Celular , Divisão Celular , Células Cultivadas , Dexametasona/farmacologia , Hepatócitos/citologia , Masculino , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , alfa-Fetoproteínas/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA