Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
3.
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
4.
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
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
6.
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
7.
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
8.
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
9.
Diabetologia ; 51(7): 1181-91, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18438639

RESUMO

AIMS/HYPOTHESIS: Based on mouse study findings, pancreatic islet cells are supposed to lack basement membrane (BM) and interact directly with vascular endothelial BM. Until now, the BM composition of human islets has remained elusive. METHODS: Immunohistochemistry with specific monoclonal and polyclonal antibodies as well as electron microscopy were used to study BM organisation and composition in human adult islets. Isolated islet cells and function-blocking monoclonal antibodies and recombinant soluble Lutheran peptide were further used to study islet cell adhesion to laminin (Lm)-511. Short-term cultures of islets were used to study Lutheran and integrin distribution. RESULTS: Immunohistochemistry revealed a unique organisation for human Lm-511/521 as a peri-islet BM, which co-invaginated into islets with vessels, forming an outer endocrine BM of the intra-islet vascular channels, and was distinct from the vascular BM that additionally contained Lm-411/421. These findings were verified by electron microscopy. Lutheran glycoprotein, a receptor for the Lm alpha5 chain, was found prominently on endocrine cells, as identified by immunohistochemistry and RT-PCR, whereas alpha(3) and beta(1) integrins were more diffusely distributed. High Lutheran content was also found on endocrine cell membranes in short-term culture of human islets. The adhesion of dispersed beta cells to Lm-511 was inhibited equally effectively by antibodies to integrin and alpha(3) and beta(1) subunits, and by soluble Lutheran peptide. CONCLUSIONS/INTERPRETATION: The present results disclose a hitherto unrecognised BM organisation and adhesion mechanisms in human pancreatic islets as distinct from mouse islets.


Assuntos
Membrana Basal/citologia , Células Endoteliais/citologia , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Adulto , Animais , Anticorpos Monoclonais , Membrana Basal/metabolismo , Membrana Basal/ultraestrutura , Biomarcadores/metabolismo , Adesão Celular , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Sistema Endócrino/citologia , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Humanos , Imuno-Histoquímica , Ilhotas Pancreáticas/ultraestrutura , Laminina/imunologia , Laminina/metabolismo , Sistema do Grupo Sanguíneo Lutheran , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Microscopia Eletrônica de Transmissão , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/metabolismo , Receptores de Laminina/imunologia , Receptores de Laminina/metabolismo
11.
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
12.
Diabetologia ; 47(2): 225-39, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14727023

RESUMO

AIMS/HYPOTHESIS: It is thought that enterovirus infections cause beta-cell damage and contribute to the development of Type 1 diabetes by replicating in the pancreatic islets. We sought evidence for this through autopsy studies and by investigating known enterovirus receptors in cultured human islets. METHODS: Autopsy pancreases from 12 newborn infants who died of fulminant coxsackievirus infections and from 65 Type 1 diabetic patients were studied for presence of enteroviral ribonucleic acid by in situ hybridisation. Forty non-diabetic control pancreases were included in the study. The expression and role of receptor candidates in cultured human islets were investigated with receptor-specific antibodies using immunocytochemistry and functional assays. RESULTS: Enterovirus-positive islet cells were found in some of both autopsy specimen collections, but not in control pancreases. No infected cells were seen in exocrine tissue. The cell surface molecules, poliovirus receptor and integrin alphavbeta3, which act as enterovirus receptors in established cell lines, were expressed in beta cells. Antibodies to poliovirus receptor, human coxsackievirus and adenovirus receptor and integrin alphavbeta3 protected islets and beta cells from adverse effects of poliovirus, coxsackie B viruses, and several of the arginine-glycine-aspartic acid motifs containing enteroviruses and human parechovirus 1 respectively. No evidence was found for expression of the decay-accelerating factor which acts as a receptor for several islet-cell-replicating echoviruses in established cell lines. CONCLUSIONS/INTERPRETATION: The results show a definite islet-cell tropism of enteroviruses in the human pancreas. Some enteroviruses seem to use previously identified cell surface molecules as receptors in beta cells, whereas the identity of receptors used by other enteroviruses remains unknown.


Assuntos
Infecções por Enterovirus/patologia , Enterovirus/crescimento & desenvolvimento , Ilhotas Pancreáticas/virologia , Receptores Virais/metabolismo , Adolescente , Adulto , Anticorpos Monoclonais/farmacologia , Autopsia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Infecções por Coxsackievirus/patologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/virologia , Echovirus 9/genética , Echovirus 9/crescimento & desenvolvimento , Enterovirus/genética , Enterovirus Humano B/genética , Enterovirus Humano B/crescimento & desenvolvimento , Humanos , Hibridização In Situ , Lactente , Recém-Nascido , Inflamação/patologia , Inflamação/virologia , Insulina/análise , Insulina/imunologia , Insulina/metabolismo , Secreção de Insulina , Integrina alfaVbeta3/análise , Integrina alfaVbeta3/imunologia , Integrina alfaVbeta3/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Proteínas de Membrana/análise , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Pessoa de Meia-Idade , Pâncreas/química , Pâncreas/patologia , Pâncreas/virologia , Parechovirus/genética , Parechovirus/crescimento & desenvolvimento , Poliovirus/genética , Poliovirus/crescimento & desenvolvimento , RNA Viral/genética , Receptores Virais/análise , Receptores Virais/imunologia
13.
Am J Physiol Endocrinol Metab ; 283(2): E207-16, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12110524

RESUMO

ATP-sensitive potassium (K(ATP)) channels are inhibited by intracellular ATP and activated by ADP. Nutrient oxidation in beta-cells leads to a rise in [ATP]-to-[ADP] ratios, which in turn leads to reduced K(ATP) channel activity, depolarization, voltage-dependent Ca(2+) channel activation, Ca(2+) entry, and exocytosis. Persistent hyperinsulinemic hypoglycemia of infancy (HI) is a genetic disorder characterized by dysregulated insulin secretion and, although rare, causes severe mental retardation and epilepsy if left untreated. The last five or six years have seen rapid advance in understanding the molecular basis of K(ATP) channel activity and the molecular genetics of HI. In the majority of cases for which a genotype has been uncovered, causal HI mutations are found in one or the other of the two genes, SUR1 and Kir6.2, that encode the K(ATP) channel. This article will review studies that have defined the link between channel activity and defective insulin release and will consider implications for future understanding of the mechanisms of control of insulin secretion in normal and diseased states.


Assuntos
Trifosfato de Adenosina/fisiologia , Doenças do Sistema Endócrino/metabolismo , Insulina/metabolismo , Canais de Potássio/metabolismo , Animais , Finlândia/epidemiologia , Humanos , Hiperinsulinismo/complicações , Hiperinsulinismo/congênito , Hiperinsulinismo/epidemiologia , Hiperinsulinismo/genética , Hipoglicemia/etiologia , Incidência , Recém-Nascido , Secreção de Insulina , Mutação , Canais de Potássio/genética
14.
Cytogenet Cell Genet ; 95(3-4): 157-62, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12063393

RESUMO

Transforming growth factor-beta (TGF-beta) superfamily related growth factors signal by binding to transmembrane type I and type II receptor serine/threonine kinases (RSTK), which phosphorylate intracellular Smad transcription factors in response to ligand binding. Here we describe the cloning of the human type I RSTK activin receptor-like kinase 7 (ALK7), an orthologue of the previously identified rat ALK7. Nodal, a TGF-beta member expressed during embryonic development and implicated in developmental events like mesoderm formation and left-right axis specification, was recently shown to signal through ALK7. We found ALK7 mRNA to be most abundantly expressed in human brain, pancreas and colon. A cDNA encoding the open reading frame of ALK7 was obtained from a human brain cDNA library. Furthermore, a P1 artificial chromosome (PAC) clone containing the human ALK7 gene was isolated and fluorescent in situ hybridization (FISH) on metaphase chromosomes identified the gene locus as chromosome 2q24.1-->q3. To test the functionality of the ALK7 signaling, we generated recombinant adenoviruses containing a constitutively active form of ALK7 (Ad-caALK7), which is capable of activating downstream targets in a ligand independent manner. Infection with Ad-caALK7 of MIN6 insulinoma cells, in which ALK7 has previously been shown to be endogenously expressed, led to a marked increase in the phosphorylation of Smad2, a signaling molecule also used by TGF-betas and activins.


Assuntos
Receptores de Ativinas Tipo I/genética , Química Encefálica/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Ativinas Tipo I/metabolismo , Sequência de Aminoácidos , Mapeamento Cromossômico , Clonagem Molecular , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Biblioteca Gênica , Humanos , Insulinoma/genética , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Proteína Smad2 , Transativadores/metabolismo , Células Tumorais Cultivadas
16.
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
17.
Genes Chromosomes Cancer ; 29(1): 83-7, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10918398

RESUMO

Comparative genomic hybridization (CGH) was used to investigate changes in DNA copy numbers in 25 paraffin-embedded samples of pancreatic endocrine tumors from 23 patients. Insulin was the dominant hormone in 12, glucagon in 7, somatostatin in 1, and pancreatic polypeptide in 2 tumors. One to 15 (mean, 8.1) changes in DNA copy numbers were observed in 22 of the 25 tumors. The most recurrent aberration, found in 68% of the tumors, involved gains in chromosome 7 with a minimal overlapping region at 7q11.2. Other frequent gains included chromosomes 19 (60%) and 14 (56%). Chromosome arm 20q was amplified in 48% of the cases with the minimal overlapping region of 20q11.1-13.1. The two most frequent DNA losses were found at 11q21-22 in 32% and at 11p13-15 in 24% of the cases. The amplified chromosomal regions contain several candidate genes that may be involved in islet cell tumorigenesis. The regions with most frequent losses are likely to contain still uncharacterized tumor suppressor genes. Wiley-Liss, Inc.


Assuntos
Aberrações Cromossômicas/genética , Neoplasias Pancreáticas/genética , Adulto , Idoso , Carcinoma de Células das Ilhotas Pancreáticas/genética , Carcinoma de Células das Ilhotas Pancreáticas/patologia , Deleção Cromossômica , Feminino , Dosagem de Genes , Glucagonoma/genética , Glucagonoma/patologia , Humanos , Insulinoma/genética , Insulinoma/patologia , Masculino , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico , Neoplasias Pancreáticas/patologia , Somatostatinoma/genética , Somatostatinoma/patologia
18.
Mol Cell Endocrinol ; 165(1-2): 41-9, 2000 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-10940482

RESUMO

AR42J is an exocrine pancreatic cell line that has been reported to differentiate towards an endocrine phenotype when stimulated with various growth factors, such as activin A, hepatocyte growth factor (HGF), betacellulin or glucagon-like peptide 1. In our experiments, AR42J-B13 cells differentiated morphologically in response to the growth factor treatment as reported previously. However, they failed to express the insulin gene. We found that the cells did not express several transcription factors known to be found in the beta-cell, including Nkx6.1, isl-1, Pax4 and Pax6. In addition, the mRNA level for pdx-1 and Nkx2.2 were very low in comparison to the insulinoma cell lines INS-1 and RINm5F. However, some transcription factors typically found in beta-cells and neuroendocrine cells were expressed also in the AR42J-B13 cells. These included BETA2/NeuroD, HNF1alpha, C/EBPbeta and IA-1. Unlike the insulinoma cells, AR42J cells expressed the exocrine transcription factor p48. In order to induce endocrine differentiation, we transfected the AR42J-B13 cells with the full length cDNAs of isl-1, Nkx6.1, Nkx2.2 and pdx-1 under the control of the CMV promoter, both separately and in combinations. The expression of Nkx2.2 led consistently to the appearance of pancreatic polypeptide but not insulin, glucagon or somatostatin mRNA. The PP mRNA expression in Nkx2.2 cDNA transfected cells was independent of the growth factor treatment used for differentiating AR42J cells. In conclusion, the AR42J-B13 line possesses some features of a pancreatic neuroendocrine cell. However, we were unable to confirm the capacity of these cells to differentiate into insulin-producing cells. Our results indicate that Nkx2.2 plays a role in the transcriptional regulation of PP expression.


Assuntos
Hormônios/biossíntese , Pâncreas/metabolismo , Fatores de Transcrição/genética , Ativinas , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Primers do DNA/genética , DNA Complementar/genética , Expressão Gênica/efeitos dos fármacos , Glucagon/biossíntese , Glucagon/genética , Fator de Crescimento de Hepatócito/farmacologia , Proteína Homeobox Nkx-2.2 , Hormônios/genética , Inibinas/farmacologia , Insulina/biossíntese , Insulina/genética , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Polipeptídeo Pancreático/biossíntese , Polipeptídeo Pancreático/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Somatostatina/biossíntese , Somatostatina/genética , Transfecção
19.
Development ; 127(12): 2617-27, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10821760

RESUMO

Pancreatic acini and islets are believed to differentiate from common ductal precursors through a process requiring various growth factors. Epidermal growth factor receptor (EGF-R) is expressed throughout the developing pancreas. We have analyzed here the pancreatic phenotype of EGF-R deficient (-/-) mice, which generally die from epithelial immaturity within the first postnatal week. The pancreata appeared macroscopically normal. The most striking feature of the EGF-R (-/-) islets was that instead of forming circular clusters, the islet cells were mainly located in streak-like structures directly associated with pancreatic ducts. Based on BrdU-labelling, proliferation of the neonatal EGF-R (-/-) beta-cells was significantly reduced (2.6+/-0.4 versus 5.8+/-0.9%, P<0.01) and the difference persisted even at 7-11 days of age. Analysis of embryonic pancreata revealed impaired branching morphogenesis and delayed islet cell differentiation in the EGF-R (-/-) mice. Islet development was analyzed further in organ cultures of E12.5 pancreata. The proportion of insulin-positive cells was significantly lower in the EGF-R (-/-) explants (27+/-6 versus 48+/-8%, P<0.01), indicating delayed differentiation of the beta cells. Branching of the epithelium into ducts was also impaired. Matrix metalloproteinase (MMP-2 and MMP-9) activity was reduced 20% in EGF-R (-/-) late-gestation pancreata, as measured by gelatinase assays. Furthermore, the levels of secreted plasminogen activator inhibitor-1 (PAI-1) were markedly higher, while no apparent differences were seen in the levels of active uPA and tPa between EGF-R (-/-) and wild-type pancreata. Our findings suggest that the perturbation of EGF-R-mediated signalling can lead to a generalized proliferation defect of the pancreatic epithelia associated with a delay in beta cell development and disturbed migration of the developing islet cells as they differentiate from their precursors. Upregulated PAI-1 production and decreased gelatinolytic activity correlated to this migration defect. An intact EGF-R pathway appears to be a prerequisite for normal pancreatic development.


Assuntos
Receptores ErbB/fisiologia , Ilhotas Pancreáticas/embriologia , Animais , Apoptose , Glicemia/metabolismo , Diferenciação Celular , Movimento Celular , Desenvolvimento Embrionário e Fetal , Receptores ErbB/deficiência , Receptores ErbB/genética , Idade Gestacional , Ilhotas Pancreáticas/citologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Morfogênese , Pâncreas/citologia , Pâncreas/embriologia , Fenótipo
20.
Diabetes ; 48(2): 408-15, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10334322

RESUMO

Mutations in genes encoding the ATP-regulated potassium (K(ATP)) channels of the pancreatic beta-cell (SUR1 and Kir6.2) are the major known cause of persistent hyperinsulinemic hypoglycemia of infancy (PHHI). We collected all cases of PHHI diagnosed in Finland between 1983 and 1997 (n = 24). The overall incidence was 1:40,400, but in one area of Central Finland it was as high as 1:3,200. Haplotype analysis using polymorphic markers spanning the SUR1/Kir6.2 gene cluster confirmed linkage to the 11p region. Sequence analysis revealed a novel point mutation in exon 4 of SUR1, predicting a valine to aspartic acid change at amino acid 187 (V187D). Of the total cases, 15 affected individuals harbored this mutation in heterozygous or homozygous form, and all of these had severe hyperinsulinemia that responded poorly to medical treatment and required subtotal pancreatectomy. No K(ATP) channel activity was observed in beta-cells isolated from a homozygous patient or after coexpression of recombinant Kir6.2 and SUR1 carrying the V187D mutation. Thus, the mutation produces a nonfunctional channel and, thereby, continuous insulin secretion. This unique SUR1 mutation explains the majority of PHHI cases in Finland and is strongly associated with a severe form of the disease. These findings provide diagnostic and prognostic utility for suspected PHHI patients.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Hiperinsulinismo/complicações , Hiperinsulinismo/genética , Hipoglicemia/etiologia , Hipoglicemia/genética , Mutação Puntual/genética , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/genética , Receptores de Droga/genética , Trifosfato de Adenosina/fisiologia , Animais , Eletrofisiologia , Feminino , Finlândia , Haplótipos/genética , Humanos , Incidência , Lactente , Recém-Nascido , Ilhotas Pancreáticas/metabolismo , Masculino , Mutação/genética , Canais de Potássio/metabolismo , Canais de Potássio/fisiologia , Proteínas Recombinantes , Receptores de Sulfonilureias , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA