Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
BMC Med ; 20(1): 495, 2022 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-36581877

RESUMO

BACKGROUND: Prenatal alcohol exposure (PAE) affects embryonic development, causing a variable fetal alcohol spectrum disorder (FASD) phenotype with neuronal disorders and birth defects. We hypothesize that early alcohol-induced epigenetic changes disrupt the accurate developmental programming of embryo and consequently cause the complex phenotype of developmental disorders. To explore the etiology of FASD, we collected unique biological samples of 80 severely alcohol-exposed and 100 control newborns at birth. METHODS: We performed genome-wide DNA methylation (DNAm) and gene expression analyses of placentas by using microarrays (EPIC, Illumina) and mRNA sequencing, respectively. To test the manifestation of observed PAE-associated DNAm changes in embryonic tissues as well as potential biomarkers for PAE, we examined if the changes can be detected also in white blood cells or buccal epithelial cells of the same newborns by EpiTYPER. To explore the early effects of alcohol on extraembryonic placental tissue, we selected 27 newborns whose mothers had consumed alcohol up to gestational week 7 at maximum to the separate analyses. Furthermore, to explore the effects of early alcohol exposure on embryonic cells, human embryonic stem cells (hESCs) as well as hESCs during differentiation into endodermal, mesodermal, and ectodermal cells were exposed to alcohol in vitro. RESULTS: DPPA4, FOXP2, and TACR3 with significantly decreased DNAm were discovered-particularly the regulatory region of DPPA4 in the early alcohol-exposed placentas. When hESCs were exposed to alcohol in vitro, significantly altered regulation of DPPA2, a closely linked heterodimer of DPPA4, was observed. While the regulatory region of DPPA4 was unmethylated in both control and alcohol-exposed hESCs, alcohol-induced decreased DNAm similar to placenta was seen in in vitro differentiated mesodermal and ectodermal cells. Furthermore, common genes with alcohol-associated DNAm changes in placenta and hESCs were linked exclusively to the neurodevelopmental pathways in the enrichment analysis, which emphasizes the value of placental tissue when analyzing the effects of prenatal environment on human development. CONCLUSIONS: Our study shows the effects of early alcohol exposure on human embryonic and extraembryonic cells, introduces candidate genes for alcohol-induced developmental disorders, and reveals potential biomarkers for prenatal alcohol exposure.


Assuntos
Transtornos do Espectro Alcoólico Fetal , Proteínas Nucleares , Efeitos Tardios da Exposição Pré-Natal , Feminino , Humanos , Recém-Nascido , Gravidez , Biomarcadores/metabolismo , Cromatina , Deficiências do Desenvolvimento , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Placenta/metabolismo
2.
Diabetologia ; 56(1): 31-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23073708

RESUMO

AIMS/HYPOTHESIS: Exercise-induced hyperinsulinism (EIHI) is a hypoglycaemic disorder characterised by inappropriate insulin secretion following anaerobic exercise or pyruvate load. Activating promoter mutations in the MCT1 gene (also known as SCLA16A1), coding for monocarboxylate transporter 1 (MCT1), were shown to associate with EIHI. Recently, transgenic Mct1 expression in pancreatic beta cells was shown to introduce EIHI symptoms in mice. To date, MCT1 has not been demonstrated in insulin-producing cells from an EIHI patient. METHODS: In vivo insulin secretion was studied during an exercise test before and after the resection of an insulinoma. The presence of MCT1 was analysed using immunohistochemistry followed by laser scanning microscopy, western blot analysis and real-time RT-PCR of MCT1. The presence of MCT1 protein was analysed in four additional insulinoma patients. RESULTS: Clinical testing revealed massive insulin secretion induced by anaerobic exercise preoperatively, but not postoperatively. MCT1 protein was not detected in the patient's normal islets. In contrast, immunoreactivity was clearly observed in the insulinoma tissue. Western blot analysis and real-time RT-PCR showed a four- to fivefold increase in MCT1 in the insulinoma tissue of the EIHI patient compared with human pancreatic islets. MCT1 protein was detected in three of four additional insulinomas. CONCLUSIONS/INTERPRETATION: We show for the first time that an MCT1-expressing insulinoma was associated with EIHI and that MCT1 might be present in most insulinomas. Our data suggest that MCT1 expression in human insulin-producing cells can lead to EIHI and warrant further studies on the role of MCT1 in human insulinoma patients.


Assuntos
Hiperinsulinismo/etiologia , Hipoglicemia/etiologia , Células Secretoras de Insulina/metabolismo , Insulinoma/fisiopatologia , Transportadores de Ácidos Monocarboxílicos/metabolismo , Atividade Motora , Proteínas de Neoplasias/metabolismo , Simportadores/metabolismo , Adolescente , Teste de Esforço , Feminino , Humanos , Hiperinsulinismo/fisiopatologia , Hipoglicemia/prevenção & controle , Células Secretoras de Insulina/patologia , Insulinoma/metabolismo , Insulinoma/patologia , Insulinoma/cirurgia , Masculino , Pessoa de Meia-Idade , Transportadores de Ácidos Monocarboxílicos/genética , Fases do Sono , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/prevenção & controle , Simportadores/genética , Resultado do Tratamento , Inconsciência/etiologia , Inconsciência/prevenção & controle
3.
J Virol ; 86(8): 4463-7, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301147

RESUMO

Fibroblasts can be reprogrammed into induced pluripotent stem cells (iPSC) by ectopic expression of key transcription factors. Current methods for the generation of integration-free iPSC are limited by the low efficiency of iPSC generation and by challenges in reprogramming methodology. Recombinant adeno-associated virus (rAAV) is a potent gene delivery vehicle capable of efficient transduction of transgenic DNA into cells. rAAV stays mainly as an episome in nondividing cells, and the extent of integration is still poorly defined for various replicating cells. In this study, we aimed to induce iPSC from mouse and human fibroblasts by using rAAV vector-mediated transient delivery of reprogramming factors. We succeeded in deriving induced pluripotent stem cells from mouse but not human fibroblasts. Unexpectedly, the rAAV vector-mediated reprogramming led to frequent genomic integration of vector sequences during the reprogramming process, independent of the amount of virus used, and to persistent expression of reprogramming factors in generated iPSC clones. It thus appears that rAAV vectors are not compatible with the derivation of integration-free iPSC.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução Genética , Integração Viral , Animais , Diferenciação Celular , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Pediatr Diabetes ; 13(2): 176-80, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21595807

RESUMO

OBJECTIVE: We compared the prevalence and characteristics of diabetes between Somali and Finnish children in the City of Helsinki. SUBJECTS AND METHODS: Ten Somali and 310 non-Somali children <16 yr of age were treated for diabetes in Helsinki at the end of 2007. We analyzed autoantibodies, HLA alleles, and serum 25-hydroxy-vitamin D [S25(OH)D] concentrations. RESULTS: The prevalence of diabetes was 40/10,000 (95% CI 19-73/10,000) for the Somali children and 37/10,000 (95% CI 33-41/10,000) for the background population. At least one autoantibody was detected in all seven Somali patients sampled within 18 months after the diagnosis. Most Somalis (75%) carried HLA-conferred susceptibility to type 1 diabetes (T1D), DR3-DQ2 being the dominating HLA haplotype. Low S25(OH)D levels (<40 nmol/L) were seen in 83% of the Somali patients and in 60% of their siblings. CONCLUSIONS: These data show that (i) Somali children have autoimmune diabetes, (ii) the prevalence of T1D is similar among Somali and Finnish children, and (iii) both affected and unaffected Somali children have low concentrations of S25(OH)D.


Assuntos
População Negra/estatística & dados numéricos , Diabetes Mellitus Tipo 1/etnologia , Adolescente , Autoanticorpos/sangue , Autoanticorpos/imunologia , População Negra/genética , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Feminino , Finlândia/epidemiologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Lactente , Masculino , Prevalência , Somália/etnologia , Vitamina D/análogos & derivados , Vitamina D/sangue
5.
Diabetologia ; 54(7): 1735-43, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21509441

RESUMO

AIMS/HYPOTHESIS: Epidermal growth factor receptor (EGFR) signalling is essential for the proper fetal development of pancreatic islets and in the postnatal formation of an adequate beta cell mass. In this study we investigated the role of EGFR signalling in the physiological states of beta cell mass expansion in adults during metabolic syndrome and pregnancy, as well as in regeneration after pancreatic duct ligation. METHODS: Heterozygous Pdx1-EGFR-dominant-negative (E1-DN) mice, which have a kinase-negative EGFR under the Pdx1 promoter, and wild-type mice were both subjected to a high-fat diet, pregnancy and pancreatic duct ligation. RESULTS: The beta cell mass of wild-type mice fed the high-fat diet increased by 70% and the mice remained normoglycaemic; the E1-DN mice became diabetic and failed to show any compensatory beta cell mass expansion. Similarly, pregnant wild-type mice had four times more proliferating beta cells and a 75% increase in beta cell mass at mid-gestation, in contrast to the pregnant E1-DN mice, which did not show any significant beta cell compensation and were hyperglycaemic in an intraperitoneal glucose tolerance test. However, after pancreatic duct ligation, both the wild-type and E1-DN mice showed similar expression of Ngn3 (also known as Neurog3) and beta cell proliferation increased to a similar level in the ligated part of pancreas. CONCLUSIONS/INTERPRETATIONS: EGFR signalling is essential in beta cell mass expansion during a high-fat diet and pregnancy where replication is the primary mechanism for compensatory beta cell mass expansion. In contrast, EGFR signalling appears not to be crucial to increased beta cell proliferation after pancreatic duct ligation.


Assuntos
Gorduras na Dieta/efeitos adversos , Receptores ErbB/metabolismo , Células Secretoras de Insulina/patologia , Ligadura/efeitos adversos , Ductos Pancreáticos/lesões , Animais , Receptores ErbB/genética , Feminino , Imuno-Histoquímica , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Diabetologia ; 52(4): 621-5, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19169662

RESUMO

AIMS/HYPOTHESIS: Isolated pure human beta cells would be helpful for a number of research purposes. However, lack of beta cell-specific surface antigens has been a major problem. We aimed to develop a simple method for human beta cell isolation based on the initial elimination of ductal cells by their expression of carbohydrate antigen 19-9 (CA19-9), followed by positive selection of beta cells by their expression of polysialic acid-neural cell adhesion molecule (PSA-NCAM). METHODS: Cell type-specific expression of CA19-9, NCAM and PSA-NCAM was studied in sections of adult human pancreas and in cultured primary endocrine and exocrine cells. Dispersed human islet cells were purified in two steps, after 4 days of suspension culture, by binding to magnetic microbeads coupled to antibodies against CA19-9 and PSA-NCAM. RESULTS: NCAM expression was detected in ducts and islets in the human pancreas. In contrast, PSA-NCAM immunoreactivity was detected only in islets. PSA-NCAM staining in dispersed cells revealed that the marker is expressed in all endocrine cell types, but not in duct cells. Purification of dispersed islet cells using PSA-NCAM microbeads alone did not completely eliminate contaminating duct cells. However, elimination of the duct cells by CA19-9 microbeads followed by positive sorting of the PSA-NCAM-positive cells in five consecutive islet preparations resulted in 90 to 98% pure endocrine cells, of which 89 to 97% were beta cells. CONCLUSIONS/INTERPRETATION: We describe a simple and reproducible method for purification of viable human pancreatic beta cells devoid of exocrine acini and ducts.


Assuntos
Separação Celular/métodos , Células Secretoras de Insulina/citologia , Antígeno CA-19-9/análise , Ácido Edético , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/fisiologia , Molécula L1 de Adesão de Célula Nervosa/análise , Moléculas de Adesão de Célula Nervosa/análise , Ductos Pancreáticos/citologia , Ácidos Siálicos/análise , Tripsina
8.
Clin Endocrinol (Oxf) ; 70(1): 96-103, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19128366

RESUMO

OBJECTIVE: Homozygous mutations in the gene encoding the pituitary transcription factor PROP1 are associated with combined pituitary hormone deficiency (CPHD) in both mice and humans with a highly variable phenotype with respect to the severity and time of initiation of pituitary hormone deficiency. We have ascertained three pedigrees with PROP1 mutations from a large cohort of patients with variable degrees of CPHD who were screened for mutations in PROP1. RESULTS: Affected individuals from all three pedigrees were found to harbour novel PROP1 mutations. We have identified two siblings in one family who were homozygous for an intronic mutation (c.343-11C > G) that disrupts correct splicing resulting in the loss of exon 3 from the PROP1 transcript. Two siblings from a second, unrelated family are compound heterozygotes for two point mutations in the coding region, a missense mutation (p.R125W) that leads to impaired transcriptional activation, and a deletion of a single nucleotide (c.310delC) resulting in a frameshift and nonfunctional mutant protein. Additionally, we identified a homozygous deletion of the PROP1 locus in two patients born to consanguineous parents. CONCLUSION: Mutations in PROP1 are a frequent cause of familial CPHD. We have described four novel mutations in PROP1 in 3 pedigrees, all resulting in PROP1 deficiency by different mechanisms. The phenotypic variation observed in association with PROP1 mutations both within and between families, together with the evolving nature of hormone deficiencies and sometimes changing pituitary morphology indicates a need for continual monitoring of these patients.


Assuntos
Proteínas de Homeodomínio/genética , Hipopituitarismo/genética , Hormônios Hipofisários/deficiência , Adolescente , Animais , Células CHO , Criança , Pré-Escolar , Estudos de Coortes , Cricetinae , Cricetulus , Análise Mutacional de DNA , Feminino , Deleção de Genes , Humanos , Lactente , Masculino , Linhagem
9.
Diabetes Obes Metab ; 10 Suppl 4: 119-27, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18834439

RESUMO

Basement membranes (BMs) are an important part of the physiological microenvironment of pancreatic islet cells. In mouse islets, beta-cells interact directly with BMs of capillary endothelial cells. We have shown that in the human islets, the capillaries are surrounded by a double BM both in foetal and adult tissues. The endocrine islet cells are facing a BM that is separate from the endothelia. Laminins are the functionally most important component of BMs. The only laminin isoform present in the human endocrine islet BM is laminin-511 (previously known as laminin 10). The islet cells facing this BM have a strong and polarized expression of Lutheran glycoprotein, which is a well-known receptor for the laminin alpha 5 chain. Dispersed human islet cells adhere to purified human laminin-511 and the binding is equally effectively blocked by a soluble form of Lutheran as by antibody against integrin beta1. Our results reveal unique features of the BM structure of human islets, different from rodents. This information has potentially important implications for the generation of an optimal microenvironment for beta-cell function, proliferation and differentiation.


Assuntos
Membrana Basal/fisiologia , Diferenciação Celular/fisiologia , Matriz Extracelular/fisiologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiopatologia , Laminina/fisiologia , Pâncreas/fisiopatologia , Animais , Membrana Basal/embriologia , Membrana Basal/metabolismo , Ciclo Celular/fisiologia , Matriz Extracelular/metabolismo , Humanos , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Laminina/metabolismo , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Camundongos , Pâncreas/embriologia , Isoformas de Proteínas/metabolismo , Receptores de Laminina/metabolismo
10.
Horm Res ; 70(2): 65-72, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18547951

RESUMO

In recent years, considerable progress has been made in the biochemical, morphological and molecular genetic differentiation of congenital hyperinsulinism (CHI). Fluorine-18 L-3,4-dihydroxyphenylalanine positron emission tomography ((18)F-DOPA-PET) has been introduced for differentiation between focal and diffuse CHI. The ability to take up L-DOPA and convert it into dopamine is correlated with the activity of the aromatic amino acid decarboxylase and increased in the hyperfunctional affected pancreatic area in comparison to normally functioning pancreas. The high sensitivity of this method allows the surgeon to perform a curative limited resection of a focus without the risk of long-term diabetes. The exact preoperative planning by (18)F-DOPA-PET/CT computer tomography allows laparoscopic operation in selected cases with the focus in the tail and limits necessity to open the pancreatic duct in cases with focus in the head. Patients with persistent CHI should be managed within a strong network of diagnostic, treatment, and research institutions.


Assuntos
Hiperinsulinismo Congênito/diagnóstico , Di-Hidroxifenilalanina , Radioisótopos de Flúor , Tomografia por Emissão de Pósitrons , Algoritmos , Hiperinsulinismo Congênito/cirurgia , Humanos , Pâncreas/cirurgia , Cuidados Pré-Operatórios
11.
J Clin Invest ; 92(3): 1459-66, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8104197

RESUMO

The effects of nicotinamide (NIC) on human fetal and adult endocrine pancreatic cells were studied in tissue culture. Treatment of the fetal cells with 10 mM NIC resulted in a twofold increase in DNA content and a threefold increase in insulin content. This was associated with the development of beta cell outgrowths from undifferentiated epithelial cell clusters and an increase in the expression of the insulin, glucagon, and somatostatin genes. DNA synthesis was stimulated only in the undifferentiated cells. Half-maximal doses for the insulinotropic and mitogenic effects of NIC were 5-10 and 1-2 mM, respectively. Islet-like cell clusters cultured with NIC responded to glucose stimulation with a biphasic increase in insulin release (fourfold peak), whereas control cells were unresponsive to glucose. Both control and NIC-treated cells developed into functional islet tissue after transplantation into athymic nude mice. As compared with adult islets, the insulinotropic action of NIC could only be demonstrated in the fetal cells. Our results indicate that NIC induces differentiation and maturation of human fetal pancreatic islet cells. This model should be useful for the study of molecular mechanisms involved in beta cell development.


Assuntos
Ilhotas Pancreáticas/citologia , Niacinamida/farmacologia , Pâncreas/embriologia , Adulto , Fatores Etários , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cultura , Expressão Gênica , Glucagon/genética , Humanos , Insulina/genética , Inibidores de Poli(ADP-Ribose) Polimerases , RNA Mensageiro/genética , Somatostatina/genética
12.
J Clin Invest ; 106(7): 897-906, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11018078

RESUMO

ATP-sensitive potassium channels play a major role in linking metabolic signals to the exocytosis of insulin in the pancreatic beta cell. These channels consist of two types of protein subunit: the sulfonylurea receptor SUR1 and the inward rectifying potassium channel Kir6.2. Mutations in the genes encoding these proteins are the most common cause of congenital hyperinsulinism (CHI). Since 1973, we have followed up 38 pediatric CHI patients in Finland. We reported previously that a loss-of-function mutation in SUR1 (V187D) is responsible for CHI of the most severe cases. We have now identified a missense mutation, E1506K, within the second nucleotide binding fold of SUR1, found heterozygous in seven related patients with CHI and in their mothers. All patients have a mild form of CHI that usually can be managed by long-term diazoxide treatment. This clinical finding is in agreement with the results of heterologous coexpression studies of recombinant Kir6.2 and SUR1 carrying the E1506K mutation. Mutant K(ATP) channels were insensitive to metabolic inhibition, but a partial response to diazoxide was retained. Five of the six mothers, two of whom suffered from hypoglycemia in infancy, have developed gestational or permanent diabetes. Linkage and haplotype analysis supported a dominant pattern of inheritance in a large pedigree. In conclusion, we describe the first dominantly inherited SUR1 mutation that causes CHI in early life and predisposes to later insulin deficiency.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Genes Dominantes , Hiperinsulinismo/congênito , Mutação de Sentido Incorreto , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/genética , Receptores de Droga/genética , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Adolescente , Adulto , Sítios de Ligação , Glicemia/metabolismo , Criança , Pré-Escolar , Diazóxido/farmacologia , Diazóxido/uso terapêutico , Feminino , Finlândia , Humanos , Hiperinsulinismo/tratamento farmacológico , Masculino , Pâncreas/patologia , Linhagem , Polimorfismo Conformacional de Fita Simples , Canais de Potássio/efeitos dos fármacos , Receptores de Droga/efeitos dos fármacos , Receptores de Sulfonilureias , Tolbutamida/farmacologia
13.
J Leukoc Biol ; 78(4): 845-52, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16037409

RESUMO

Macrophages are a heterogeneous population of cells that belong to the mononuclear phagocyte system. They play an important role in tissue homeostasis and remodeling and are also potent immune regulators. Pancreatic macrophages are critically involved in the development and pathogenesis of autoimmune diabetes. To elucidate the ontogeny of pancreatic macrophages, we characterized in this study the macrophages present in the adult and developing fetal pancreas of normal mice. We additionally examined the presence of local macrophage precursors and the involvement of macrophages in the growth of endocrine tissue in the fetal pancreas. We identified two phenotypically distinct macrophage subsets in the adult pancreas. The majority of macrophages was CD45(+)ER-MP23(+)MOMA-1(+). Under noninflammatory conditions, only a minority ( approximately 5%) of the pancreatic macrophages additionally expressed the macrophage marker F4/80. In contrast, in the fetal pancreas, phenotypically, mature macrophages were identified exclusively by their expression of F4/80 and lacked detectable staining with ER-MP23 and MOMA-1 antibodies. In fetal pancreas organ cultures, we could show that macrophages develop from pre-existing precursors, which are present in the fetal pancreas at embryonic age 12.5. Moreover, the number of macrophages increased significantly when macrophage-colony stimulating factor was added to these cultures. It is important that this increase of F4/80-positive cells was paralleled by an increase in the number of insulin-producing cells, suggesting that macrophages support the growth of these endocrine cells.


Assuntos
Sistema Endócrino/embriologia , Macrófagos/citologia , Macrófagos/imunologia , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Animais , Antígenos de Diferenciação/imunologia , Linhagem da Célula/imunologia , Sistema Endócrino/imunologia , Feminino , Técnicas In Vitro , Células Secretoras de Insulina/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pâncreas/imunologia , Fenótipo
14.
Diabetes ; 43(9): 1164-6, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8070617

RESUMO

Reg is a gene associated with regeneration of pancreatic islets. We have previously shown that nicotinamide induces differentiation of human fetal beta-cells in tissue culture and that hepatocyte growth factor/scatter factor (HGF/SF) is mitogenic for the fetal beta-cells. We now tested whether these conditions, supporting either differentiation or growth, are associated with changes in reg gene expression in human fetal pancreatic cells. Culture for 7 days with 10 mM nicotinamide led to a fourfold decrease in reg mRNA levels (23 +/- 12% of control, n = 5, P < 0.001). In contrast, HGF/SF increased reg expression threefold (302 +/- 68% of control, n = 4, P < 0.05). Nicotinamide, which does not alter the differentiation level of adult beta-cells, did not significantly affect reg expression in adult human islets (84 +/- 4% of control, n = 2, NS). Thus, a higher level of endocrine differentiation is associated with a lower level of reg expression, and a higher rate of beta-cell replication results in increased reg transcription. These results provide the first evidence of a molecular marker, the reg gene, to distinguish between proliferation and differentiation of human beta-cells.


Assuntos
Proteínas de Ligação ao Cálcio/biossíntese , Expressão Gênica , Fator de Crescimento de Hepatócito/farmacologia , Ilhotas Pancreáticas/citologia , Proteínas do Tecido Nervoso , Isomerases de Aminoácido/biossíntese , Proteínas de Transporte/biossíntese , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Endodesoxirribonucleases/biossíntese , Feto , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Litostatina , Niacinamida/farmacologia , Peptidilprolil Isomerase , Fosfoproteínas/biossíntese , RNA Mensageiro/biossíntese , Transcrição Gênica
15.
Diabetes ; 37(3): 286-91, 1988 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-3286329

RESUMO

The insulin release in response to glucose was studied in perifused isletlike cell clusters (ICCs) obtained from human fetal or neonatal pancreases at various stages of development: 12-15 gestational wk (n = 7), 17-20 wk (n = 13), 22.5 wk (n = 2, 1 diabetic pregnancy), and 26-44 wk (n = 6, postnatal samples). The ICCs were stimulated with 20 mM glucose and subsequently with 10 mM theophylline plus 20 mM glucose as a viability test. Insulin release increased to a detectable level (greater than 0.1 pg.ICC-1.min-1) during glucose stimulation in four of seven of the youngest fetuses. At 17-20 wk the basal rate of insulin release had increased by at least 15-fold above the detection limit (1.5 pg.ICC-1.min-1), and glucose promoted a sustained monophasic response that was on the average 1.6-fold higher than the basal level. The response was significant (P less than .05) in 9 of 13 experiments. With postnatal ICC (gestational age 26-44 wk), an early-phase peak response was observed in 5 of 6 experiments. The mean rates of insulin release after 5-12 min of glucose stimulation were 4.8 pg.ICC-1.min-1 in newborn infants and 2.1 pg.ICC-1.min-1 in 17- to 20-wk fetuses. The corresponding mean relative insulin responses (stimulated to basal) were 3.3-fold (range 1.1-7.5) and 1.6-fold (1.0-3.4), respectively (P less than .05, Mann-Whitney U test). The results suggest that the human fetal pancreas is already responsive to glucose during the first half of gestation, but the biphasic insulin release does not start to mature until the postnatal phase.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Pâncreas/metabolismo , Células Cultivadas , Desenvolvimento Embrionário e Fetal , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Insulina/uso terapêutico , Ilhotas Pancreáticas/metabolismo , Neonatologia , Pâncreas/efeitos dos fármacos , Gravidez , Gravidez em Diabéticas/tratamento farmacológico , Estatística como Assunto
16.
Diabetes ; 37(12): 1678-83, 1988 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-3056761

RESUMO

The effects of recombinant human growth hormone (GH, 1 micrograms/ml) and insulin-like growth factor I (IGF-I, 200 ng/ml) on the production of insulin and glucagon by human fetal islet-like cell clusters (ICCs) were studied in tissue culture. ICCs were derived after collagenase digestion and culture of pancreases from 16 fetuses (mean gestational age 15.6 wk). The ICCs were cultured with or without GH or IGF-I for 7 or 31 days. Basal rates of insulin and glucagon production were not altered by GH during the first 17 days of culture, but the release of both hormones was increasingly augmented by GH during the last 2 wk of culture (131% increase in insulin and 85% in glucagon compared with controls). ICCs cultured for 7 days in the presence of GH secreted more insulin when incubated for 120 min in 20 mM than in 2 mM glucose (2.1-fold response, P less than .05), whereas ICCs maintained in basal medium did not respond to glucose. GH had no effect on DNA and insulin content or insulin biosynthesis. Exogenous IGF-I caused a 28% suppression of insulin release (P less than .05) between days 4 and 10 of culture but induced a 49% increase in the mean secretion rate during the last week (days 25-31, P less than .01). Glucagon release was not affected by exogenous IGF-I. In contrast to GH, exogenous IGF-I induced a twofold increase in the DNA content of the 7-day--cultured ICCs. However, insulin biosynthesis and release were markedly suppressed.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Feto/efeitos dos fármacos , Glucagon/metabolismo , Hormônio do Crescimento/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/embriologia , Somatomedinas/farmacologia , Técnicas de Cultura , DNA/análise , Feto/fisiologia , Humanos , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos
17.
Diabetes ; 50(2): 301-7, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11272140

RESUMO

Neogenesis is crucial for the maintenance of beta-cell mass in the human pancreas and possibly for the outcome of clinical islet transplantation. To date, no studies have reported a stimulation of human beta-cell neogenesis in vivo. Therefore, we investigated whether human alpha-, beta-, and duct cell growth can be stimulated when human islets are xenotransplanted to obese hyperglycemic-hyperinsulinemic ob/ob mice immunosuppressed with anti-lymphocyte serum. Moreover, we wanted to study whether beta-cell growth and duct-to-beta-cell differentiation were induced in the hepatocyte growth factor (HGF)-dependent compensatory kidney growth model. For that purpose, we evaluated human islets grafted to nude (nu/nu) mice before uninephrectomy of the contralateral kidney for DNA-synthesis and duct cell expression of the beta-cell-specific transcription factor Nkx 6.1 as an estimate of differentiation. Human islet grafts were well preserved after 2 weeks when transplanted to ob/ob mice during anti-lymphocyte immunosuppression. Both human beta-cells (P < 0.01) and duct cells (P < 0.001) were growth stimulated when islets were transplanted to ob/ob mice. We also observed a correlation between increased duct cell proliferation and increased organ donor age (P = 0.02). Moreover, duct (P < 0.05) and beta-cell (P < 0.05) proliferation, as well as duct cell Nkx 6.1 expression (P < 0.05), were enhanced by the compensatory kidney growth after uninephrectomy. We conclude that it is possible to stimulate human beta-cell neogenesis in vivo, provided that the recipient carries certain growth-stimulatory traits. Furthermore, it seems that duct cell proliferation increases with increasing organ donor age. Altogether, these data and previous results from our laboratory suggest that human beta-cell neogenesis becomes more dependent on differentiation and less dependent on proliferation with increasing age.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/citologia , Leptina/genética , Transplante Heterólogo , Adaptação Fisiológica , Adolescente , Adulto , Animais , Diferenciação Celular , Divisão Celular , Criança , Feminino , Humanos , Terapia de Imunossupressão , Ilhotas Pancreáticas/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL/genética , Pessoa de Meia-Idade , Nefrectomia , Suínos
18.
Diabetes ; 46(2): 244-8, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9000701

RESUMO

The scarcity of human adult islets available for transplantation in IDDM makes the use of human fetal pancreatic cells desirable. Human fetal pancreatic cells grow and differentiate after transplantation in nude mice. It is unclear whether proliferation of preexisting endocrine cells or differentiation of precursor cells is mainly responsible for the increased islet mass and if beta-cell enrichment before transplantation enhances the functional outcome of the graft. To answer these questions, we transplanted purified human fetal islets, islet-like cell clusters (ICCs), and fresh tissue under the kidney capsule of nude mice. Insulin content was highest in the fresh tissue but fell rapidly during culture as either fetal islets or ICCs. Although fetal islets contained fourfold more insulin than ICCs before transplantation, the insulin content of the resulting grafts was the same after 3 months in vivo. The degree of stimulation after glucose challenge was comparable; however, more tissue was needed to generate the fetal islets. Grafts of fresh tissue also had similar total insulin contents, but when normalized to DNA, insulin concentration was significantly higher in the grafts from cultured tissue. Moreover, there were distinct morphological differences; the grafts from fresh tissue were more fibrous, with prominent ductal and cystic elements. Grafts from cultured tissue were two- to threefold enriched in endocrine tissue when compared with grafts originating from fresh tissue. These results suggest that islet cells identified in the grafted ICCs are mainly derived through differentiation of endocrine precursors and that cultured ICCs are more preferable than either fetal islets or uncultured tissue for transplantation.


Assuntos
Transplante das Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/citologia , Fatores Etários , Animais , Diferenciação Celular , Divisão Celular , Separação Celular , Células Cultivadas , Feto , Humanos , Insulina/metabolismo , Camundongos , Camundongos Nus , Transplante Heterólogo
19.
Diabetes ; 45(4): 496-501, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8603772

RESUMO

One of the major beta-cell autoantigens associated with IDDM is GAD. Although GAD expression has been detected in adult islets, transcriptional expression of the GAD genes has not been reported during human pancreatic ontogeny. We therefore analyzed patterns of GAD gene transcription by quantitating the mRNAs encoding both the 65- and 67-kDa isoforms (GAD65 and GAD67, respectively) in human fetal, postnatal, and adult pancreases, as well as in isolated adult islets, and examined their tissue-specific expression. Significant levels of pancreatic GAD65 transcripts were already detected at 13 weeks of gestation and were expressed at higher levels in the fetal and infantile pancreas than in the adult pancreas. Isolated adult pancreatic islets were highly enriched in GAD65 mRNA. In contrast, GAD67 transcripts were not detectable in fetal and postnatal pancreases. In addition to the pancreas, marked GAD expression was detected in the brain, whereas other tissues examined contained either low or undetectable GAD transcripts. Triple immunofluorescent staining of fetal and adult pancreases revealed colocalization of GAD65 with alpha- and beta-cells. In the fetal pancreas, strong immunoreactivity for GAD65 was also evident in epithelial cells, which lacked expression of insulin or glucagon, some of which were present in the ductal epithelium, suggesting that GAD65 expression might correlate with endocrine determination. In summary, 1) this is the first demonstration of GAD65 expression in the human fetal pancreas, implicating a potential role during islet development, and 2) GAD65 may be a useful marker for the identification of primitive islet cells.


Assuntos
Desenvolvimento Embrionário e Fetal , Expressão Gênica , Glutamato Descarboxilase/biossíntese , Pâncreas/enzimologia , Adulto , Encéfalo/embriologia , Encéfalo/enzimologia , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/imunologia , Feto , Imunofluorescência , Idade Gestacional , Glucagon/análise , Glutamato Descarboxilase/análise , Humanos , Imuno-Histoquímica , Insulina/análise , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/enzimologia , Especificidade de Órgãos , Pâncreas/embriologia , Transcrição Gênica
20.
Diabetes ; 43(7): 947-53, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8013761

RESUMO

Fetal mesenchyme-derived factors are likely to play an important role in pancreatic islet development and growth. We have used primary cultures of human fetal pancreatic tissue to identify growth factors that have morphogenic, mitogenic, and insulinotropic activity. The formation of islet-like cell clusters (ICCs) during a 6-day culture was stimulated two- to threefold by hepatocyte growth factor/scatter factor (HGF/SF) basic fibroblast growth factor (FGF)-2, and to a lesser extent by keratinocyte growth factor (FGF-7) and insulin-like growth factor-II (IGF-II). In contrast, transforming growth factor-beta (TGF-beta) had a strong inhibitory effect. The ICCs formed during HGF/SF stimulation consisted mainly of epithelial cells, whereas FGF-2-induced ICCs were predominantly nonepithelial. Furthermore, although both FGF-2 and HGF/SF increased the total insulin content of the cultures, only HGF/SF increased the insulin content per DNA. Quantitatively, HGF/SF stimulated a 2.3-fold increase in the proportion of insulin-positive cells and a 3-fold higher number of replicating beta-cells. Blocking of the IGF-I receptor inhibited ICC formation but did not affect their insulin content. Immunoneutralizing TGF-beta resulted in increased cell growth and insulin content, indicating the presence of an endogenous inhibitory TGF-beta activity in the model system. Our results suggest that HGF/SF may be an important component of the fetal mesenchyme-derived factors responsible for pancreatic islet development. HGF/SF also may prove valuable for supporting the in vitro growth of islet cells.


Assuntos
Substâncias de Crescimento/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Pâncreas/embriologia , Aborto Legal , Análise de Variância , Anticorpos/farmacologia , Anticorpos Monoclonais/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , DNA/metabolismo , Feminino , Feto , Idade Gestacional , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/embriologia , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Gravidez , Receptor IGF Tipo 1/imunologia , Receptor IGF Tipo 1/fisiologia , Timidina/metabolismo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA