Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Stem Cells ; 27(8): 1941-53, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19544426

RESUMO

The success of cell replacement therapy for diabetes depends on the availability and generation of an adequate number of islets, preferably from an autologous origin. Stem cells are now being probed for the generation of physiologically competent, insulin-producing cells. In this investigation, we explored the potential of adipose tissue-derived stem cells (ASCs) to differentiate into pancreatic hormone-expressing islet-like cell aggregates (ICAs). We initiated ASC culture from epididymal fat pads of Swiss albino mice to obtain mesenchymal cells, murine epididymal (mE)-ASCs. Subsequent single-cell cloning resulted in a homogeneous cell population with a CD29(+)CD44(+)Sca-1(+) surface antigen expression profile. We formulated a 10-day differentiation protocol to generate insulin-expressing ICAs from mE-ASCs by progressively changing the differentiation cocktail on day 1, day 3, and day 5. Our stage-specific approach successfully differentiated mesodermic mE-ASCs into definitive endoderm (cells expressing Sox17, Foxa2, GATA-4, and cytokeratin [CK]-19), then into pancreatic endoderm (cells expressing pancreatic and duodenal homeobox [PDX]-1, Ngn3, NeuroD, Pax4, and glucose transporter 2), and finally into cells expressing pancreatic hormones (insulin, glucagon, somatostatin). Fluorescence-activated cell sorting analysis showed that day 5 ICAs contained 64.84% +/- 7.03% PDX-1(+) cells, and in day 10 mature ICAs, 48.17% +/- 3% of cells expressed C-peptide. Day 10 ICAs released C-peptide in a glucose-dependent manner, exhibiting in vitro functionality. Electron microscopy of day 10 ICAs revealed the presence of numerous secretory granules within the cell cytoplasm. Calcium alginate-encapsulated day 10 ICAs (1,000-1,200), when transplanted i.p. into streptozotocin-induced diabetic mice, restored normoglycemia within 2 weeks. The data presented here demonstrate the feasibility of using ASCs as a source of autologous stem cells to differentiate into the pancreatic lineage.


Assuntos
Tecido Adiposo/citologia , Ilhotas Pancreáticas/citologia , Hormônios Pancreáticos/biossíntese , Células-Tronco/citologia , Animais , Agregação Celular/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Diabetes Mellitus Experimental/terapia , Endoderma/metabolismo , Epididimo , Regulação da Expressão Gênica , Insulina/metabolismo , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos , Estreptozocina
2.
Stem Cells ; 25(12): 3215-22, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17901402

RESUMO

Islet transplantation offers improved glucose homeostasis in diabetic patients, but transplantation of islets is limited by the supply of donor pancreases. Undifferentiated precursors hold promise for cell therapy because they can expand before differentiation to produce a large supply of functional insulin-producing cells. Previously, we described proliferative populations of human islet-derived precursor cells (hIPCs) from adult islets. To show the differentiation potential of hIPCs, which do not express insulin mRNA after at least 1,000-fold expansion, we generated epithelial cell clusters (ECCs) during 4 days of differentiation in vitro. After transplantation into mice, 22 of 35 ECC preparations differentiated and matured into functional cells that secreted human C-peptide in response to glucose. Transcripts for insulin, glucagon, and somatostatin in recovered ECC grafts increased with time in vivo, reaching levels approximately 1% of those in adult islets. We show that hIPCs are mesenchymal stromal cells (MSCs) that adhere to plastic, express CD73, CD90, and CD105, and can differentiate in vitro into adipocytes, chondrocytes, and osteocytes. Moreover, we find a minor population of CD105(+)/CD73(+)/CD90(+) cells in adult human islets (prior to incubation in vitro) that express insulin mRNA at low levels. We conclude that hIPCs are a specific type of pancreas-derived MSC that are capable of differentiating into hormone-expressing cells. Their ability to mature into functional insulin-secreting cells in vivo identifies them as an important adult precursor or stem cell population that could offer a virtually unlimited supply of human islet-like cells for replacement therapy in type 1 diabetes. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Ilhotas Pancreáticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Hormônios Pancreáticos/biossíntese , Células Estromais/citologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Humanos , Insulina/biossíntese , Insulina/genética , Ilhotas Pancreáticas/citologia , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Hormônios Pancreáticos/genética , Células Estromais/metabolismo
3.
J Immunol ; 178(5): 3007-15, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17312146

RESUMO

The prevalent view of thymic epithelial differentiation and Aire activity holds that Aire functions in terminally differentiated medullary thymic epithelial cells (MTECs) to derepress the expression of structural tissue-restricted Ags, including pancreatic endocrine hormones. An alternative view of these processes has proposed that Aire functions to regulate the differentiation of immature thymic epithelial cells, thereby affecting tissue-restricted Ag expression and negative selection. In this study, we demonstrate that Aire impacts several aspects of murine MTECs and provide support for this second model. Expression of transcription factors associated with developmental plasticity of progenitor cells, Nanog, Oct4, and Sox2, by MTECs was Aire dependent. Similarly, the transcription factors that regulate pancreatic development and the expression of pancreatic hormones are also expressed by wild-type MTECs in an Aire-dependent manner. The altered transcriptional profiles in Aire-deficient MTECs were accompanied by changes in the organization and composition of the medullary epithelial compartment, including a reduction in the medullary compartment defined by keratin (K) 14 expression, altered patterns of K5 and K8 expression, and more prominent epithelial cysts. These findings implicate Aire in the regulation of MTEC differentiation and the organization of the medullary thymic compartment and are compatible with a role for Aire in thymic epithelium differentiation.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Timo/imunologia , Fatores de Transcrição/imunologia , Animais , Diferenciação Celular/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Queratinas/biossíntese , Queratinas/imunologia , Camundongos , Camundongos Mutantes , Pâncreas/crescimento & desenvolvimento , Pâncreas/imunologia , Pâncreas/metabolismo , Hormônios Pancreáticos/biossíntese , Hormônios Pancreáticos/imunologia , Timo/metabolismo , Timo/patologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
J Histochem Cytochem ; 55(1): 97-104, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16982847

RESUMO

The desert gerbil Psammomys obesus, an established model of type 2 diabetes (T2D), has previously been shown to lack pancreatic and duodenal homeobox gene 1 (Pdx-1) expression. Pdx-1 deficiency leads to pancreas agenesis in both mice and humans. We have therefore further examined the pancreas of P. obesus during embryonic development. Using Pdx-1 antisera raised against evolutionary conserved epitopes, we failed to detect Pdx-1 immunoreactivity at any time points. However, at E14.5, Nkx6.1 immunoreactivity marks the nuclei of all epithelial cells of the ventral and dorsal pancreatic buds and the only endocrine cell types found at this time point are glucagon and PYY. At E18.5 the pancreas is well branched and both glucagon- and ghrelin-positive cells are scattered or found in clusters, whereas insulin-positive cells are not found. At E22.5, the acini of the exocrine pancreas are starting to mature, and amylase and carboxypeptidase A immunoreactivity is found scattered and not in all acini. Ghrelin-, glucagon-, PYY-, gastrin-, somatostatin (SS)-, pancreatic polypeptide (PP)-, and insulin-immunoreactive cells are found scattered or in small groups within or lining the developing ductal epithelium as marked by cytokeratin 19. Using degenerate PCR, the P. obesus Neurogenin-3 (Ngn-3) gene was cloned. Nucleotide and amino acid sequences show high homology with known Ngn-3 sequences. Using specific antiserum, we can observe that Ngn-3-immunoreactive cells are rare at E14.5 but readily detectable at E18.5 and E22.5. In conclusion, despite the lack of detection of Pdx-1, the P. obesus pancreas develops similarly to Muridae species, and the Ngn-3 sequence and expression pattern is highly conserved in P. obesus.


Assuntos
Pâncreas/metabolismo , Hormônios Pancreáticos/biossíntese , Fatores de Transcrição/biossíntese , Animais , Clonagem Molecular , Gerbillinae , Idade Gestacional , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Pâncreas/embriologia , Hormônios Pancreáticos/genética , Ratos , Especificidade da Espécie , Fatores de Transcrição/genética
5.
Nat Biotechnol ; 24(11): 1392-401, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17053790

RESUMO

Of paramount importance for the development of cell therapies to treat diabetes is the production of sufficient numbers of pancreatic endocrine cells that function similarly to primary islets. We have developed a differentiation process that converts human embryonic stem (hES) cells to endocrine cells capable of synthesizing the pancreatic hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, gut-tube endoderm, pancreatic endoderm and endocrine precursor--en route to cells that express endocrine hormones. The hES cell-derived insulin-expressing cells have an insulin content approaching that of adult islets. Similar to fetal beta-cells, they release C-peptide in response to multiple secretory stimuli, but only minimally to glucose. Production of these hES cell-derived endocrine cells may represent a critical step in the development of a renewable source of cells for diabetes cell therapy.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Células Enteroendócrinas/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Hormônios Pancreáticos/biossíntese , Hormônios Peptídicos/biossíntese , Células Cultivadas , Grelina , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Pâncreas/citologia , Hormônios Pancreáticos/isolamento & purificação
6.
Lab Invest ; 84(5): 607-17, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15034596

RESUMO

Recent findings suggest that bone marrow (BM) cells have the capacity to differentiate into a variety of cell types including endocrine cells of the pancreas. We report that BM derived cells, when cultured under defined conditions, were induced to trans-differentiate into insulin-producing cells. Furthermore, these insulin-producing cells formed aggregates that, upon transplantation into mice, acquired architecture similar to islets of Langerhans. These aggregates showed endocrine gene expression for insulin (I and II), glucagon, somatostatin and pancreatic polypeptide. Immunohistochemistry also confirmed that these aggregates were positive for insulin, somatostatin, pancreatic polypeptide and C-peptide. Also, Western and ELISA analysis demonstrated expression of proinsulin and/or secretion of active insulin upon glucose challenge. Subcapsular renal transplantation of these aggregates into hyperglycemic mice lowered circulating blood glucose levels and maintained comparatively normal glucose levels for up to 90 days post-transplantation. Graft removal resulted in rapid relapse and death in experimental animals. In addition, electron microscopy revealed these aggregates had acquired ultrastructure typically associated with mature beta (beta) cells. These results demonstrate that adult BM cells are capable of trans-differentiating into a pancreatic lineage in vitro and may represent a pool of cells for the treatment of diabetes mellitus.


Assuntos
Transplante de Medula Óssea , Diabetes Mellitus Tipo 1/terapia , Insulina/biossíntese , Transplante das Ilhotas Pancreáticas , Animais , Transplante de Medula Óssea/patologia , Transplante de Medula Óssea/fisiologia , Diferenciação Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Expressão Gênica , Insulina/genética , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia Eletrônica , Hormônios Pancreáticos/biossíntese , Hormônios Pancreáticos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Transplante Heterólogo
7.
Regul Pept ; 107(1-3): 63-9, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12137967

RESUMO

OBJECTIVES: Ghrelin, an endogenous ligand of the growth hormone secretagogue receptor (GHS-R), was recently identified in the stomach. Ghrelin is produced in a population of endocrine cells in the gastric mucosa, but expression in intestine, hypothalamus and testis has also been reported. Recent data indicate that ghrelin affects insulin secretion and plays a direct role in metabolic regulation and energy balance. On the basis of these findings, we decided to examine whether ghrelin is expressed in human pancreas. Specimens from fetal to adult human pancreas and stomach were studied by immunocytochemistry, for ghrelin and islet hormones, and in situ hybridisation, for ghrelin mRNA. RESULTS: We identified ghrelin expression in a separate population of islet cells in human fetal, neonatal, and adult pancreas. Pancreatic ghrelin cells were numerous from midgestation to early postnatally (10% of all endocrine cells). The cells were few, but regularly seen in adults as single cells at the islet periphery, in exocrine tissue, in ducts, and in pancreatic ganglia. Ghrelin cells did not express any of the known islet hormones. In fetuses, at midgestation, ghrelin cells in the pancreas clearly outnumbered those in the stomach. CONCLUSIONS: Ghrelin is expressed in a quite prominent endocrine cell population in human fetal pancreas, and ghrelin expression in the pancreas precedes by far that in the stomach. Pancreatic ghrelin cells remain in adult islets at lower numbers. Ghrelin is not co-expressed with any known islet hormone, and the ghrelin cells may therefore constitute a new islet cell type.


Assuntos
Ilhotas Pancreáticas/citologia , Pâncreas/metabolismo , Hormônios Peptídicos/biossíntese , Adulto , Contagem de Células , Mucosa Gástrica/metabolismo , Grelina , Humanos , Imuno-Histoquímica , Hibridização In Situ , Recém-Nascido , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Microscopia de Fluorescência , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Hormônios Pancreáticos/biossíntese , RNA Mensageiro/biossíntese
8.
Regul Pept ; 83(2-3): 123-7, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10511466

RESUMO

The purpose of this study was to test the hypothesis that the endoprotease, prohormone convertase-1 (PC-1), is involved in the processing of the precursor protein chromogranin A (CGA) to a smaller peptide called pancreastatin (PST). A human pancreatic carcinoid cell line (BON) that expresses PC-1, CGA and PST was stably transfected with antisense PC-1 mRNA. BON cells expressing antisense PC-1 mRNA showed nearly complete abolishment of PC-1 protein (approximately 95% reduction) and an 80% reduction in cell content of PST immunoreactivity (PST-IR) as assessed by high-performance liquid chromatography in combination with measurement of PST-IR. These findings indicate that PC-1 is essential for processing CGA to PST.


Assuntos
Ácido Aspártico Endopeptidases/fisiologia , Cromograninas/metabolismo , Hormônios Pancreáticos/metabolismo , Ácido Aspártico Endopeptidases/genética , Tumor Carcinoide/enzimologia , Tumor Carcinoide/metabolismo , Cromatografia Líquida de Alta Pressão , Cromogranina A , Humanos , Hormônios Pancreáticos/biossíntese , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/metabolismo , Pró-Proteína Convertases , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas
9.
Neuroimmunomodulation ; 6(1-2): 108-14, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-9876241

RESUMO

The thymus plays a central role in the selection of T lymphocytes that are tolerant to 'self' antigens and responsive to foreign pathogens. We and others have reported the expression of the pancreatic endocrine hormones, preproinsulin, proglucagon, prosomatostatin and propancreatic polypeptide in the human and mouse thymus. While mRNA expression is very low there is evidence for the presence of the translated product. In addition, we have investigated the cell types responsible for expression. In the thymus, hormone expression is enriched in the antigen-presenting cell population. Interestingly, while proglucagon, prosomatostatin and propancreatic polypeptide appear to be expressed in a macrophage population, preproinsulin expression was restricted to dendritic cells which are more potent antigen-presenting cells. The functional significance of the endogenous expression of insulin in the thymus has been indirectly investigated using transgenic models in which the transgene is introduced by the rat insulin promoter. The data suggest that thymic expression of the transgene is critical in the induction of T-cell tolerance to the transgene in the periphery. Taken together, the evidence suggests that the low-level pancreatic hormone expression in the thymus may be involved in central tolerance to proteins of restricted expression.


Assuntos
Hormônios Pancreáticos/biossíntese , Precursores de Proteínas/biossíntese , Timo/fisiologia , Animais , Deleção Clonal , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Humanos , Tolerância Imunológica , Linfócitos T/imunologia
10.
Microsc Res Tech ; 43(4): 313-21, 1998 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9849972

RESUMO

Understanding of islet embryogenesis may prove to be key in the design of future therapies for diabetes directed at re-initiating islet growth, with the goal to replace and/or replenish the impaired beta-cell mass in the disease. In this context, studies of islet neurohormonal peptides, known to play a role in the local regulation of islet function, and their expression during islet embryogenesis are important. Here we review our studies on the embryonic islet expression of islet amyloid polypeptide (IAPP) and the PP-fold peptides pancreatic polypeptide (PP), peptide YY (PYY) and neuropeptide Y (NPY). IAPP, which is constitutively expressed in beta- and delta-cells in the adult rat, was found to occur in the assumed pluripotent islet progenitor cell, together with PYY, glucagon, and to a lesser extent with insulin. As development proceeds, the insulin/IAPP phenotype is segregated from that of PYY/glucagon; with the formation of islet-like structures, insulin/IAPP-expressing cells primarily occupy their central portions, while PYY/glucagon-expressing cells are found in their periphery. At the time of formation of islet-like structures, expression of NPY is induced in the insulin/IAPP-containing cells. Whereas NPY-expression ceases at birth, PYY is constitutively expressed in non-beta-cells in the mature rat. Expression of PP is induced just prior to birth in a separate population of islet cells, occasionally co-expressed with PYY. Although a clear role for these peptides during embryogenesis has not been identified, they conceivably could play a role in the control of insulin secretion, islet growth and islet blood flow.


Assuntos
Amiloide/biossíntese , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Neuropeptídeo Y/biossíntese , Polipeptídeo Pancreático/biossíntese , Peptídeo YY/análise , Amiloide/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Neuropeptídeo Y/genética , Pâncreas/embriologia , Hormônios Pancreáticos/biossíntese , Polipeptídeo Pancreático/genética , Peptídeo YY/genética , Ratos
11.
Mol Cell Endocrinol ; 140(1-2): 45-50, 1998 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-9722167

RESUMO

Ontogenic relationships between the different endocrine cell types of the islets of Langerhans were explored by generating transgenic mice, in which cells transcribing the glucagon, insulin, or pancreatic polypeptide genes were destroyed through the promoter-targeted expression of the diphtheria toxin A chain. In an alternate approach, to assess whether insulin cells are derived from precursors producing glucagon or PP, transgenic mice were generated bearing an insulin promoter-driven, and loxP-containing ('floxed') reporter transgene that can be irreversibly 'tagged' by recombination. They were crossed with mice expressing another transgene ('tagger') encoding Cre (cyclization recombination) recombinase in either glucagon or PP cells. The results obtained using both approaches indicate that neither glucagon nor insulin gene-expressing cells are the precursors to the other islet cells; also, they suggest that PP gene-expressing cells are necessary for the differentiation of islet insulin and somatostatin cells, through a cell lineage or a paracrine relationship.


Assuntos
Linhagem da Célula , Ilhotas Pancreáticas/citologia , Transgenes/genética , Proteínas Virais , Animais , Diferenciação Celular , Toxina Diftérica/genética , Toxina Diftérica/toxicidade , Regulação da Expressão Gênica , Genes Reporter/genética , Integrases/genética , Ilhotas Pancreáticas/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Hormônios Pancreáticos/biossíntese , Hormônios Pancreáticos/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/toxicidade , Recombinação Genética , Sequências Reguladoras de Ácido Nucleico , Células-Tronco/citologia
12.
Horm Metab Res ; 29(6): 271-7, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9230348

RESUMO

Pancreatic islets of Langerhans exhibit an architecture and cellular organization ideal for rapid, yet finely controlled, responses to changes in blood glucose levels. In type I, insulin-dependent diabetes (IDD), this organization is lost as a result of the progressive autoimmune response which selectively destroys the insulin-producing pancreatic beta cells. Since beta cells are perceived as end-stage differentiated cells having limited capacity for regeneration in situ, individuals with IDD resulting from beta cell loss or dysfunction require life-long insulin therapy. Efforts to produce islet neogenesis or initiate islet growth in vitro from either fetal or adult tissue have had minimal success. We now report that pancreatic-derived, pluripotent islet-producing stem cells (IPSCs), isolated from prediabetic mice, can be grown in long-term cultures and differentiated into immature functional islet-like structures containing cells which express low levels of insulin, glucagon and/or somatostatin. When such in vitro grown islets were implanted into clinically diabetic NOD mice, the implanted mice were successfully weaned from insulin long-term (>50 days) without ill effects. The implanted mice maintained blood glucose levels just above euglycemic (180-220 mg/dl) and showed no signs of disease. Thus, this technical breakthrough provides new therapeutic approaches to diabetes as an alternative to insulin therapy.


Assuntos
Ilhotas Pancreáticas/fisiologia , Pâncreas/citologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Primers do DNA , Hormônios/biossíntese , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Endogâmicos NOD , Microscopia Eletrônica de Varredura , Oligonucleotídeos Antissenso , Hormônios Pancreáticos/biossíntese , Hormônios Pancreáticos/genética , Fenótipo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
13.
Regul Pept ; 70(2-3): 121-33, 1997 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-9272624

RESUMO

The ECL cells constitute the predominant endocrine cell population in the mucosa of the acid-secreting part of the stomach (fundus). They are rich in chromogranin A (CGA), histamine and histidine decarboxylase (HDC). They secrete CGA-derived peptides and histamine in response to gastrin. The objective of this investigation was to examine the expression of pancreastatin (rat CGA266-314) and WE14 (rat CGA343-356) in rat stomach ECL cells. The distribution and cellular localisation of pancreastatin- and WE14-like immunoreactivities (LI) were analysed by radioimmunoassay and immunohistochemistry with antibodies against pancreastatin, WE14 and HDC. The effect of food deprivation on circulating pancreastatin-LI was examined in intact rats and after gastrectomy or fundectomy. Rats received gastrin-17 (5 nmol/kg/h) by continuous intravenous infusion or omeprazole (400 micromol/kg) once daily by the oral route, to induce hypergastrinemia. CGA-derived peptides in the ECL cells were characterised by gel permeation chromatography. The expression of CGA mRNA was examined by Northern blot analysis. Among all of the endocrine cells in the body, the ECL cell population was the richest in pancreastatin-LI, containing 20-25% of the total body content. Food deprivation and/or surgical removal of the ECL cells lowered the level of pancreastatin-LI in serum by about 80%. Activation of the ECL cells by gastrin infusion or omeprazole treatment raised the serum level of pancreastatin-LI, lowered the concentrations of pancreastatin- and WE14-LI in the ECL cells and increased the CGA mRNA concentration. Chromatographic analysis of the various CGA immunoreactive components in the ECL cells of normal and hypergastrinemic rats suggested that these cells respond to gastrin with a preferential release of the low-molecular-mass forms.


Assuntos
Cromograninas/metabolismo , Mucosa Gástrica/metabolismo , Proteínas de Neoplasias/biossíntese , Hormônios Pancreáticos/biossíntese , Animais , Cromogranina A , Cromograninas/química , Cromograninas/genética , Células Enterocromafins , Mucosa Gástrica/química , Mucosa Gástrica/citologia , Gastrinas/sangue , Imuno-Histoquímica , Masculino , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Hormônios Pancreáticos/sangue , Hormônios Pancreáticos/química , Hormônios Pancreáticos/genética , RNA Mensageiro/química , Ratos , Ratos Sprague-Dawley
14.
Regul Pept ; 68(3): 165-75, 1997 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-9100283

RESUMO

It has been suggested that members of the neuropeptide Y (NPY) family of regulatory peptides [NPY, peptide YY (PYY) and pancreatic polypeptide (PP)] play an important role in the development of the endocrine pancreas. The development of rat endocrine pancreas from embryonic (E) day 12 until 30 days postpartum (P) was studied with emphasis on NPY, PYY and PP and their co-existence with insulin, glucagon and somatostatin using single and double immunostaining and in situ hybridization. Already at E12, PYY was detectable in small endocrine cell clusters and found to be co-localised with both insulin and glucagon, which at this stage occurred in the same cells. At E16 most of the insulin-immunoreactive (IR) cells were distinct from the glucagon/PYY-IR cells. Interestingly, at E16 NPY mRNA, and at E17 NPY immunoreactivity appeared in a few, scattered endocrine cells. Virtually all NPY-IR endocrine cells were insulin-producing beta cells. At E18 the endocrine cells started to form typical islets with centrally located insulin/NPY-IR cells surrounded by glucagon/PYY-IR cells. AT E20-E21, the vast majority of insulin-producing cells also expressed NPY. However, at birth (day 0) islet cell NPY mRNA was lacking. Postnatally the number and immunostaining intensity of NPY-IR islet cells rapidly declined, being non-detectable at P5. Cells containing PP immunoreactivity and PP mRNA were first detected at E21. The adult pattern of islet peptide distribution, with NPY confined to neuronal elements. PYY and PP exclusively in endocrine cells, was established at P5. The beta cell expression of NPY during the latter part of embryogenesis coincides with the prepartal glucocorticoid surge and with rapid islet cell replication and differentiation. This is compatible with steroid induction of NPY expression and with a role for NPY in the maturation of beta cells and their hormone release, which occurs in the immediate neonatal period.


Assuntos
Ilhotas Pancreáticas/embriologia , Neuropeptídeo Y/biossíntese , Hormônios Pancreáticos/biossíntese , Polipeptídeo Pancreático/biossíntese , Biossíntese Peptídica , Animais , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Glucagon/análise , Hibridização In Situ , Insulina/análise , Ilhotas Pancreáticas/inervação , Ilhotas Pancreáticas/metabolismo , Masculino , Neuropeptídeo Y/análise , Neuropeptídeo Y/genética , Hormônios Pancreáticos/genética , Polipeptídeo Pancreático/genética , Peptídeo YY , Peptídeos/genética , Precursores de Proteínas/análise , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Somatostatina/análise
15.
J Mol Endocrinol ; 16(1): 1-8, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8672228

RESUMO

Pancreastatin is a 49 amino acid peptide first isolated, purified and characterized from the porcine pancreas, and whose biological activity in different tissues can be assigned to the C-terminal part of the molecule. Pancreastatin has a prohormonal precursor, chromogranin A (CGA), which is a glycoprotein present in neuroendocrine cells, including the endocrine pancreas. Both intracellular and extracellular processing of CGA can yield pancreastatin. This processing is tissue-specific, with the pancreatic islet and antral gastric endocrine cells being the major source of fully processed pancreastatin. Most of the circulating CGA is secreted by chromaffin tissue. Therefore, peripheral processing of CGA is probably the major indirect source of pancreastatin. Pancreastatin seems to have a general modulatory control on endocrine (insulin, glucagon, parathormone) and exocrine (pancreatic, gastric) secretion from tissues close to the source of production. This has led to the assumption that pancreastatin may be a peptide with an autocrine and paracrine function. It has recently been revealed to be a peptide with a metabolic function counter-regulatory to insulin action. This effect, in conjunction with the inhibitory effect on insulin and pancreatic exocrine secretion, points to a role in the physiology of stress. The molecular mechanism of the glycogenolytic effect of pancreastatin is better known, although further work is still needed. In general, more studies should be carried out at the molecular level to investigate the mechanism of action of pancreastatin and thus to clarify its physiological role in the neuroendocrine system.


Assuntos
Hormônios Pancreáticos/fisiologia , Animais , Cromogranina A , Cromograninas/metabolismo , Glândulas Endócrinas/fisiologia , Glândulas Exócrinas/fisiologia , Humanos , Ilhotas Pancreáticas/fisiologia , Sistemas Neurossecretores/fisiologia , Pâncreas/fisiologia , Hormônios Pancreáticos/biossíntese , Suínos
16.
Endocrinology ; 136(5): 2252-61, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7720675

RESUMO

Brief phorbol ester treatment of BON cells results in a persistent release and cellular depletion of immunoreactive chromogranin A (CGA-IR) and neurotensin (NT-IR) cell contents. The purpose of the present study was to characterize the effects of 12-O-tetradecanoyl phorbol-13-acetate (TPA) on the secretion, biosynthesis, and steady-state messenger RNA (mRNA) levels of chromogranin A (CGA) and of a coresident peptide, neurotensin, by a novel human pancreatic carcinoid cell line, called BON. Acute TPA treatment (100 nM, 1 h) of BON cells resulted in 20- and 40-fold elevations in release of CGA-IR and NT-IR, respectively; and a 70-90% depletion of CGA-IR and NT-IR cell contents. TPA treatment also increased the biosynthetic rate of CGA-IR. Steady-state mRNA levels of CGA and NT/N (neurotensin/neuromedin N) were unchanged. Cell contents of CGA-IR and NT-IR were not replenished for a period of up to 6 days; secretion of CGA-IR and NT-IR persisted. In addition, BON cells failed to release CGA in response to stimulation by ionomycin and A23187 several days after acute TPA treatment. Our data indicate that the lack of replenishment of cell contents of CGA-IR and NT-IR is not due to decreases in steady-state CGA-IR and NT-IR mRNA levels, nor is it due to a decrease in biosynthesis of CGA-IR, but it is the result of a loss in the ability of TPA-treated BON cells to store and secrete CGA-IR and NT-IR in a regulated manner. These effects of TPA are mediated through the PKC pathway.


Assuntos
Cromograninas/biossíntese , Expressão Gênica/efeitos dos fármacos , Neurotensina/biossíntese , Análise de Variância , Northern Blotting , Calcimicina/farmacologia , Tumor Carcinoide , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cromogranina A , Cromograninas/metabolismo , Relação Dose-Resposta a Droga , Humanos , Ionomicina/farmacologia , Cinética , Metionina/metabolismo , Neurotensina/metabolismo , Hormônios Pancreáticos/biossíntese , Hormônios Pancreáticos/metabolismo , Neoplasias Pancreáticas , Fragmentos de Peptídeos/biossíntese , RNA Mensageiro/metabolismo , Radioisótopos de Enxofre , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
18.
Regul Pept ; 52(3): 195-203, 1994 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-7800852

RESUMO

Chromogranin A (CGA) is thought to be a precursor of pancreastatin (PST). Carbachol (Cch) stimulated the secretion of CGA and PST from QGP-1N cells derived from a human pancreatic islet cell tumor. Atropine inhibited the secretion of both. Sodium fluoride, phorbol ester, and calcium ionophore also stimulated the secretion of both. Cch (10(-5) M) stimulated inositol 1,4,5-trisphosphate production in QGP-1N cells. Stimulation with Cch increased the total amount of PST in the cells and the medium 1.7-fold and decreased the amount of CGA in the cells and medium. QGP-1N cells were labelled with [35S]methionine, and then CGA and PST in the cells and medium were immunoprecipitated with specific antisera, and separated by electrophoresis in polyacrylamide gel. Stimulation with Cch resulted in an increase in the intensity of PST-immunoreactive bands and a decrease in those of CGA-immunoreactive bands. Cch did not increase the cellular level of CGA messenger RNA. These results suggested that (1) the secretion of CGA and PST from QGP-1N cells is regulated mainly through muscarinic receptors coupled with activation of polyphosphoinositide breakdown by a G protein, with intracellular calcium ion and protein kinase C playing a role in the stimulus-secretion coupling and that (2) Cch may induce the secretion of PST and CGA and processing from CGA to PST.


Assuntos
Carcinoma de Células das Ilhotas Pancreáticas/fisiopatologia , Cromograninas/fisiologia , Hormônios Pancreáticos/fisiologia , Neoplasias Pancreáticas/fisiopatologia , Receptores Muscarínicos/efeitos dos fármacos , Cromogranina A , Cromograninas/biossíntese , Cromograninas/metabolismo , Meios de Cultura , Humanos , Hormônios Pancreáticos/biossíntese , Hormônios Pancreáticos/metabolismo , RNA Mensageiro/biossíntese , Estimulação Química , Células Tumorais Cultivadas
19.
Gen Comp Endocrinol ; 94(2): 244-60, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-7926634

RESUMO

The hormones of the endocrine pancreas are believed to play an important role in early development. The development of the pancreas and the appearance of hormone-producing cells during embryogenesis have been extensively studied in mammals and birds. Relatively little work has been done in other vertebrates, and there are no published studies regarding the order Crocodilia. Given the pivotal phylogenetic position of crocodilians, Alligator mississippiensis provides an interesting species in which to study the embryonic development of the endocrine pancreas. The aims of the present study were (1) to investigate the morphological development of the pancreas and (2) to determine the initial appearance and regional distribution of the pancreatic endocrine cells in the embryonic alligator. At each stage of development serial sections of pancreatic tissue were stained with hematoxylin and eosin to aid in morphological description. Using immunocytochemistry sections were stained to detect the presence of insulin, glucagon, somatostatin, and pancreatic polypeptide. The dorsal pancreatic bud was first observed at stage 8, coincident with the appearance of insulin-containing and glucagon-containing cells. Somatostatin-containing cells were first detected at stage 10. At stage 13 the ventral pancreatic bud was first observed. At stage 14 the dorsal and ventral pancreatic buds fused and insulin, glucagon, and somatostatin were found throughout the pancreas. Not until stage 17 was pancreatic polypeptide first detected. Unlike the other hormones, pancreatic polypeptide was rare or absent in the dorsal region of the pancreas. In later stages of development, somatostatin-containing cells were the most abundant and constituted 35-40% of all hormone-containing cells. The sequence of appearance of insulin and glucagon found in the alligator is the same as that found in mammals and birds.


Assuntos
Jacarés e Crocodilos/crescimento & desenvolvimento , Jacarés e Crocodilos/metabolismo , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Hormônios Pancreáticos/biossíntese , Animais , Embrião não Mamífero , Glucagon/biossíntese , Glucagon/imunologia , Imuno-Histoquímica , Insulina/biossíntese , Insulina/imunologia , Pâncreas/citologia , Hormônios Pancreáticos/imunologia , Polipeptídeo Pancreático/biossíntese , Polipeptídeo Pancreático/imunologia , Filogenia
20.
Development ; 118(4): 1031-9, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7903631

RESUMO

The early progenitor cells to the pancreatic islets in the mouse have been characterized so as to re-examine their possible lineage relationships to the four islet cell types found in mature islets. Insulin and glucagon were both first expressed at embryonic day 9.5, and many cells coexpressed these two markers, as shown by light and electron microscopic analysis using double-label immunohistochemistry. Incubation of embryonic pancreas with 1% glutaraldehyde, a fixative commonly used by electron microscopists, abolished this reactivity, thereby explaining reported difficulties in detecting these precursor cells. Using antisera specific for neuropeptide Y (NPY) a peptide with considerable homology to pancreatic polypeptide (PP), we show that NPY first appears with insulin and glucagon immunoreactivity at E9.5, and is co-expressed with glucagon in a majority of adult alpha cells. As we have previously reported, PP itself is first detectable immunocytochemically at postnatal day 1 with PP-specific antibodies. However, antibodies raised against bovine PP are shown by dot blotting to recognize NPY with comparable avidity, indicating that a recent report of islet progenitor cells containing PP at E9.5 (Herrera, P. L., Huarte, J., Sanvito, F., Meda, P., Orci, L. and Vassalli, J. D. (1991) Development 113, 1257-1265), actually represents cross-reactivity to NPY. The data support a model in which early precursor cells to the endocrine pancreas co-activate and co-express a set of islet cell hormone and neural genes, whose expression is both selectively increased and extinguished as development proceeds, concomitant with a restriction to the patterns of expression characteristic of mature islet cell types.


Assuntos
Ilhotas Pancreáticas/embriologia , Neuropeptídeo Y/biossíntese , Hormônios Pancreáticos/biossíntese , Células-Tronco/metabolismo , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Glucagon/biossíntese , Imuno-Histoquímica , Insulina/biossíntese , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Endogâmicos , Microscopia Imunoeletrônica , Polipeptídeo Pancreático/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...