Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Braz. J. Pharm. Sci. (Online) ; 58: e20065, 2022. graf
Artigo em Inglês | LILACS | ID: biblio-1403720

RESUMO

Abstract Glucose exposure induces toxic effects on the function of the pancreatic islets. Moreover, myricitrin as a flavonoid glycoside may have favorable effects on insulin secretion of Langerhans islets. The present study aimed to investigate the effect of Myricitrin and its solid lipid nanoparticles (SLN) on the insulin secretion as well as the content of isolated pancreatic islets from male mice. In this experimental study, Langerhans islets were separated from adult male NMRI mice using the collagenase method. The insulin secretion and content of islets were assessed in glucose-containing medium (2.8, 5.6, and 16.7mM). Further, islets treated were prepared by the administration of Myricitrin and its SLN (1, 3 and 10µM). Myricitrin 3µM, and SLN containing Myricitrin 3 and 10µM increased insulin secretion in medium containing glucose concentration 2.8mM. Accordingly, this variable increased in Myricitrin 3 and 10µM, SLN containing Myricitrin 1, 3, and 10µM utilization as well as glucose concentration 5.6mM. Afterward, the insulin secretion increased in medium containing 16.7mM glucose after the addition of Myricitrin and SLN containing Myricitrin 1, 3, and 10µM. Also, the insulin content increased in Myricitrin and SLN containing Myricitrin 1, 3, and 10µM administered groups in all medium containing glucose concentrations. Myricitrin and its SLN increased islets insulin secretion and content in low, moderate, and high glucose concentration mediums


Assuntos
Animais , Masculino , Camundongos , Pâncreas/efeitos dos fármacos , Ilhotas Pancreáticas/anormalidades , Secreção de Insulina/imunologia , Organização e Administração , Nanopartículas , Insulina/efeitos adversos
2.
Rev. cuba. endocrinol ; 32(2): e285, 2021. graf
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1347405

RESUMO

Introducción: El páncreas ectópico es la segunda anomalía congénita pancreática más frecuente después del páncreas divisum. Fue descrito por primera vez en 1729 por Schultz y se define como la presencia de tejido pancreático que carece de comunicación anatómica o vascular con el cuerpo principal del páncreas. La localización más frecuente es en el estómago (25 - 38 por ciento), seguido de duodeno, yeyuno e íleon. El 40 por ciento de los casos son sintomáticos y es más frecuente su presentación en varones en torno a la 5ª y 6ª década de la vida. Objetivo: Presentar un caso de páncreas ectópico diagnosticado a través de un estudio histológico tras realizada la cirugía. Presentación de caso: Presentamos el caso de una paciente compatible con hipoglucemia y cuyo estudio definitivo mostró la presencia de tejido pancreático ectópico en estómago, con resolución completa de los síntomas tras tratamiento quirúrgico. La anatomía patológica mostró una lesión nodular tumoral benigna (2,5 cm), constituida por tejido pancreático heterotópico, con presencia de páncreas exocrino con acinos. Páncreas endocrino con presencia de islotes de Langerhans y componente epitelial con ductos. Afectación desde la submucosa hasta la subserosa, con una pared muscular propia con hiperplasia muscular en relación a la heterotopía pancreática. La mucosa gástrica mostraba inflamación crónica leve con escasos folículos linfoides. Conclusiones: La presencia de páncreas ectópico es una entidad poco frecuente, pero a tener en cuenta en pacientes con clínica de hipoglucemia, una vez descartadas otras causas. No existe consenso con respecto a indicaciones en el manejo de lesiones pequeñas y asintomáticas, por lo que se recomienda individualizar cada caso teniendo en cuenta el tamaño, la localización y el tipo histológico(AU)


Introduction: Ectopic pancreas is the second most frequent congenital anomaly after pancreas divisum. It was described for the first time in 1729 by Schultz and it is defined as the presence of pancreatic tissue with no anatomical or vascular communication with the main body of pancreas. The most common location is in the stomach (25-38 percent), followed by the duodenum, jejunum and ileum ones. 40 percent of the cases are symptomatic and is more frequent their presentation in males in the fifth or sixth decade of life. Objective: To present a case of ectopic pancreas diagnosed through a histological study after surgery. Case presentation: Case of a patient with clinical features compatible with hypoglycemia that after being studied showed the presence of ectopic pancreatic tissue in the stomach, with a complete solution of the symptoms after surgical treatment. The pathological anatomy showed a benign tumor nodular lesion (2.5 cm), made up of heterotopic pancreatic tissue, with the presence of exocrine pancreas with acini. Endocrine pancreas with the presence of islets of Langerhans and epithelial component with ducts. Involvement from the submucosa to the subserosa, with a proper muscular wall with muscular hyperplasia in relation to pancreatic heterotopia. The gastric mucosa showed mild chronic inflammation with few lymphoid follicles. Conclusions: The presence of ectopic pancreas is a rare condition, but it should be taken into account in patients with clinical features of hypoglycemia once ruled out other causes. There is no consensus in regards to the indications for the management of small and asymptomatic lesions, so, it is recommended to individualize each case taking into account the size, location and histological type(AU)


Assuntos
Humanos , Feminino , Adulto , Pâncreas/anormalidades , Estômago/lesões , Ilhotas Pancreáticas/anormalidades , Hiperglicemia/etiologia
5.
Am J Pathol ; 184(5): 1446-57, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24631026

RESUMO

Prenatal and postnatal factors such as intrauterine growth restriction (IUGR) and high-fat (HF) diet contribute to type 2 diabetes. Our aim was to determine whether IUGR and HF diets interact in type 2 diabetes pathogenesis, with particular attention focused on pancreatic islet morphology including assessment for inflammation. A surgical model of IUGR (bilateral uterine artery ligation) in Sprague-Dawley rats with sham controls was used. Pups were fed either HF or chow diets after weaning. Serial measures of body weight and glucose tolerance were performed. At 25 weeks of age, rat pancreases were harvested for histologic assessment. The birth weight of IUGR pups was 13% lower than that of sham pups. HF diet caused excess weight gain, dyslipidemia, hyperinsulinemia, and mild glucose intolerance, however, this was not aggravated further by IUGR. Markedly abnormal islet morphology was evident in 0 of 6 sham-chow, 5 of 8 sham-HF, 4 of 8 IUGR-chow, and 8 of 9 IUGR-HF rats (chi-square, P = 0.007). Abnormal islets were characterized by larger size, irregular shape, inflammation with CD68-positive cells, marked fibrosis, and hemosiderosis. ß-Cell mass was not altered by IUGR. In conclusion, HF and IUGR independently contribute to islet injury characterized by inflammation, hemosiderosis, and fibrosis. This suggests that both HF and IUGR can induce islet injury via converging pathways. The potential pathogenic or permissive role of iron in this process of islet inflammation warrants further investigation.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Retardo do Crescimento Fetal/patologia , Hemossiderose/complicações , Inflamação/complicações , Inflamação/patologia , Ilhotas Pancreáticas/patologia , Animais , Glicemia/metabolismo , Peso Corporal , Dislipidemias/complicações , Jejum/sangue , Fibrose , Hemossiderose/patologia , Proteínas de Homeodomínio/metabolismo , Hiperinsulinismo/complicações , Ilhotas Pancreáticas/anormalidades , Masculino , Tamanho do Órgão , Ratos Sprague-Dawley , Transativadores/metabolismo
6.
Islets ; 2(5): 318-22, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21099330

RESUMO

The total mass of pancreatic islet cells is a critical factor in glucose metabolic control. The aim of the present study was to investigate whether in the Munich Wistar Frömter (MWF) rat, beside a reduction in the number of nephrons, there are also alterations in the number of pancreatic islets and of ß cell mass. We also examined glucose metabolism, both in normal conditions and following intravenous glucose injection. The number of islets per pancreas, estimated by morphometrical analysis, was significantly lower in MWF rats than in Wistar rats (3,501±1,285 vs. 7,259±2,330 islet/rat, respectively). Also the mean number of islets per gram of body weight was significantly lower in MWF rats than in Wistar rats (18±7 in MWF rats vs. 28±10 islets/g bw in Wistar rats). Morphometric analysis of ß cell mass showed an average of 77.1±7% islet cells staining for insulin in MWF rats and 83.9±2.1% in the control Wistar rats. Despite the lower number of islets and ß cells, MWF and Wistar rats had comparable fasting blood glucose levels but significant differences in blood glucose following an intraperitoneal glucose tolerance test. In summary, pancreatic islets of MWF and Wistar rats showed a marked difference in morphometrical characteristics. While this difference is not associated with blood glucose levels, glucose metabolism after IPGTT between MWF and Wistar rats is significantly different. These data suggest that an inborn deficit in ß cell mass of about 60% is responsible for altered glucose metabolism and could favor the development of diabetes.


Assuntos
Glucose/metabolismo , Ilhotas Pancreáticas/anormalidades , Animais , Glicemia/análise , Peso Corporal , Contagem de Células , Feminino , Intolerância à Glucose/etiologia , Teste de Tolerância a Glucose , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Tamanho do Órgão , Pâncreas/patologia , Pâncreas/fisiopatologia , Ratos , Ratos Endogâmicos
7.
FEBS Lett ; 584(4): 701-6, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20026049

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9), a liver-secreted plasma enzyme, restricts hepatic uptake of low-density lipoprotein (LDL) cholesterol by promoting the degradation of LDL receptors (LDLR). PCSK9 and LDLR are also expressed in insulin-producing pancreatic islet beta cells, possibly affecting the function of these cells. Here we show that, compared to control mice, PCSK9-null male mice over 4 months of age carried more LDLR and less insulin in their pancreas; they were hypoinsulinemic, hyperglycemic and glucose-intolerant; their islets exhibited signs of malformation, apoptosis and inflammation. Collectively, these observations suggest that PCSK9 may be necessary for the normal function of pancreatic islets.


Assuntos
Intolerância à Glucose/genética , Ilhotas Pancreáticas/metabolismo , Receptores de LDL/genética , Serina Endopeptidases/genética , Animais , Apoptose , Glicemia/metabolismo , Western Blotting , Feminino , Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/enzimologia , Teste de Tolerância a Glucose , Insulina/sangue , Insulina/metabolismo , Ilhotas Pancreáticas/anormalidades , Ilhotas Pancreáticas/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Receptores de LDL/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/deficiência , Técnicas de Cultura de Tecidos
8.
Rev Med Brux ; 31(2 Suppl): S109-12, 2010.
Artigo em Francês | MEDLINE | ID: mdl-21812222

RESUMO

Neonatal diabetes, transient (TND) or permanent (PND) is a rare disease, with a reported frequency of 1/300,000. If establishing a diagnosis is quite easy, treatment remains challenging during childhood. Understanding of physiopathology increased this last decade, as many mutations in genes playing critical roles in the development of pancreas, have been described: the most common are chromosome 6q anomalies in the case of TND, and mutations in KCNJ11 and ABCC8 genes encoding the subunit of the insulin cell potassium channel in the case of PND. We report on 2 peculiar stories: the first one is the unique case of a newborn with isodisomy of chromosome 6, methylmalonic acidemia and pancreatic beta cell agenesis, who died on the 16th day of life. The second one is the longest follow-up ever described, 38-year, of a permanent neonatal diabetes mellitus without complications, except for rare micro-aneurysms, in spite of insufficient metabolic control.


Assuntos
Diabetes Mellitus/congênito , Ilhotas Pancreáticas/anormalidades , Adulto , Aberrações Cromossômicas , Cromossomos Humanos Par 6 , Diabetes Mellitus/tratamento farmacológico , Seguimentos , Humanos , Hipoglicemiantes/uso terapêutico , Recém-Nascido , Insulina/uso terapêutico , Masculino
9.
Rev Med Brux ; 31(2 Suppl): S113-23, 2010.
Artigo em Francês | MEDLINE | ID: mdl-21812223

RESUMO

Type 2 diabetes mellitus (T2D) is no longer a disease only of adults. In some American locations and populations, incidence and prevalence of T2D are much higher than those of type 1 diabetes, because of increased calorie and fat intake, and decreased exercise. The increasing prevalence of T2D in the United States has closely paralleled the increase in childhood obesity noted there, but now across the Western world. Besides obesity, the other youth risk factors for T2D are: ethnicity, family history, puberty, female, metabolic syndrome, acanthosis nigricans and polycystic ovary syndrome. Any feature or condition associated with insulin resistance/hyperinsulinemia should alert to screen youth at increased risk for (pre)T2D. T2D should be differenciated from monogenic diabetes (Maturity Onset Diabetes of the Young or MODY). Treatment goals are to decrease weight and increase exercise, to normalize insulinemia, glycemia and HbA1c, to control hypertension and hyperlipidemia. The aim of the pharmacological therapy is to decrease insulin resistance, namely by metformin. Sometimes, insulin therapy is necessary.


Assuntos
Diabetes Mellitus Tipo 2/prevenção & controle , Adolescente , Adulto , Criança , Árvores de Decisões , Diabetes Mellitus Tipo 2/diagnóstico , Humanos , Resistência à Insulina , Ilhotas Pancreáticas/anormalidades , Obesidade/complicações , Fatores de Risco , Adulto Jovem
10.
Diabetes ; 57(9): 2421-31, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18591390

RESUMO

OBJECTIVE: Most pancreatic endocrine cells derive from Ptf1a-expressing progenitor cells. In humans, nonsense mutations in Ptf1a have recently been identified as a cause of permanent neonatal diabetes associated with pancreatic agenesis. The death of Ptf1a-null mice soon after birth has not allowed further insight into the pathogenesis of the disease; it is therefore unclear how much pancreatic endocrine function is dependent on Ptf1a in mammals. This study aims to investigate gene-dosage effects of Ptf1a on pancreas development and function in mice. RESEARCH DESIGN AND METHODS: Combining hypomorphic and null alleles of Ptf1a and Cre-mediated lineage tracing, we followed the cell fate of reduced Ptf1a-expressing progenitors and analyzed pancreas development and function in mice. RESULTS: Reduced Ptf1a dosage resulted in pancreatic hypoplasia and glucose intolerance with insufficient insulin secretion in a dosage-dependent manner. In hypomorphic mutant mice, pancreatic bud size was small and substantial proportions of pancreatic progenitors were misspecified to the common bile duct and duodenal cells. Growth with branching morphogenesis and subsequent exocrine cytodifferentiation was reduced and delayed. Total beta-cell number was decreased, proportion of non-beta islet cells was increased, and alpha-cells were abnormally intermingled with beta-cells. Interestingly, Pdx1 expression was decreased in early pancreatic progenitors but elevated to normal level at the mid-to-late stages of pancreatogenesis. CONCLUSIONS-The dosage of Ptf1a is crucial for pancreas specification, growth, total beta-cell number, islet morphogenesis, and endocrine function. Some neonatal diabetes may be caused by mutation or single nucleotide polymorphisms in the Ptf1a gene that reduce gene expression levels.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Ilhotas Pancreáticas/anormalidades , Fatores de Transcrição/genética , Animais , Tamanho Corporal , Divisão Celular/fisiologia , Duodeno/citologia , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Proteínas de Homeodomínio/genética , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Pâncreas Exócrino/anormalidades , Pâncreas Exócrino/patologia , Pâncreas Exócrino/fisiologia , Células-Tronco/citologia , Transativadores/genética
11.
Dev Dyn ; 236(10): 2865-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17849440

RESUMO

Signaling through activin type IIB receptor (ActRIIB) has been shown to regulate the axial formation and the development of foregut-derived organs such as the pancreas in mice. Here, we provide genetic evidence that ActRIIB and Smad2 genes cooperatively regulated asymmetrical patterning of the thoracic organs and pancreas development in mice. The loss of one allele of Smad2 on ActRIIB-/- background resulted in the increased severity of ActRIIB-/- phenotypes, including right pulmonary isomerism and complex cardiac malformations, and resulted in 100% frequency of death soon after birth. Of interest, 14% of compound heterozygous ActRIIB+/- Smad2+/- mice exhibited the ActRIIB-/- phenotypes and died soon after birth. In the pancreas, hypoplastic islets were found not only in ActRIIB-/- but also in Smad2+/- mice. A more severe phenotype was also found in ActRIIB+/- Smad2+/- mice. As well, these mutant mice exhibited impaired glucose tolerance in a gene dosage-sensitive manner. This genetic evidence strongly suggested that ActRIIB and Smad2 function in the same signaling pathway to regulate axial patterning and pancreas islet formation by means of a threshold mechanism.


Assuntos
Receptores de Activinas Tipo II/genética , Padronização Corporal , Coração/embriologia , Ilhotas Pancreáticas/embriologia , Pulmão/embriologia , Proteína Smad2/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Desenvolvimento Embrionário , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/anormalidades , Pulmão/anormalidades , Pulmão/metabolismo , Camundongos , Camundongos Mutantes , Pâncreas/anormalidades , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo
13.
Mol Endocrinol ; 20(8): 1853-66, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16556734

RESUMO

The FoxM1 transcription factor is highly expressed in proliferating cells and activates several cell cycle genes, although its requirement appears to be limited to certain tissue types. Embryonic hepatoblast-specific inactivation of Foxm1 results in a dramatic reduction in liver outgrowth and subsequent late gestation lethality, whereas inactivation of Foxm1 in adult liver impairs regeneration after partial hepatectomy. These results prompted us to examine whether FoxM1 functions similarly in embryonic outgrowth of the pancreas and beta-cell proliferation in the adult. We found that FoxM1 is highly expressed in embryonic and neonatal endocrine cells, when many of these cells are proliferating. Using a Cre-lox strategy, we generated mice in which Foxm1 was inactivated throughout the developing pancreatic endoderm by embryonic d 15.5 (Foxm1(Deltapanc)). Mice lacking Foxm1 in their entire pancreas were born with normal pancreatic and beta-cell mass; however, they displayed a gradual decline in beta-cell mass with age. Failure of postnatal beta-cell mass expansion resulted in impaired islet function by 6 wk of age and overt diabetes by 9 wk. The decline in beta-cell mass in Foxm1(Deltapanc) animals is due to a dramatic decrease in postnatal beta-cell replication and a corresponding increase in nuclear localization of the cyclin-dependent kinase inhibitor, p27(Kip1), a known target of FoxM1 inhibition. We conclude that Foxm1 is essential to maintain normal beta-cell mass and regulate postnatal beta-cell turnover. These results suggest that mechanisms regulating embryonic beta-cell proliferation differ from those used postnatally to maintain the differentiated cell population.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Proliferação de Células , Tamanho Celular , Regulação para Baixo , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Deleção de Genes , Expressão Gênica , Teste de Tolerância a Glucose , Insulina/biossíntese , Células Secretoras de Insulina/patologia , Integrases/metabolismo , Ilhotas Pancreáticas/anormalidades , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Pâncreas/anormalidades , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Fatores de Tempo , Fatores de Transcrição/fisiologia
14.
Mol Cell Biol ; 25(12): 4969-76, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923615

RESUMO

MafA is a transcription factor that binds to the promoter in the insulin gene and has been postulated to regulate insulin transcription in response to serum glucose levels, but there is no current in vivo evidence to support this hypothesis. To analyze the role of MafA in insulin transcription and glucose homeostasis in vivo, we generated MafA-deficient mice. Here we report that MafA mutant mice display intolerance to glucose and develop diabetes mellitus. Detailed analyses revealed that glucose-, arginine-, or KCl-stimulated insulin secretion from pancreatic beta cells is severely impaired, although insulin content per se is not significantly affected. MafA-deficient mice also display age-dependent pancreatic islet abnormalities. Further analysis revealed that insulin 1, insulin 2, Pdx1, Beta2, and Glut-2 transcripts are diminished in MafA-deficient mice. These results show that MafA is a key regulator of glucose-stimulated insulin secretion in vivo.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transativadores/metabolismo , Animais , Arginina/administração & dosagem , Arginina/metabolismo , Células Cultivadas , Diabetes Mellitus/genética , Feminino , Regulação da Expressão Gênica , Marcação de Genes , Glucose/administração & dosagem , Teste de Tolerância a Glucose , Insulina/genética , Secreção de Insulina , Ilhotas Pancreáticas/anormalidades , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Fatores de Transcrição Maf Maior , Masculino , Camundongos , Camundongos Knockout , Cloreto de Potássio/farmacologia , Transativadores/genética , Transcrição Gênica
15.
Lab Invest ; 85(1): 45-64, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15580285

RESUMO

While relatively ignored for years as vestigial, cilia have recently become the focus of intense interest as organelles that result in severe pathologies when disrupted. Here, we further establish a connection between cilia dysfunction and disease by showing that loss of polaris (Tg737), an intraflagellar transport (IFT) protein required for ciliogenesis, causes abnormalities in the exocrine and endocrine pancreas of the Tg737(orpk) mouse. Pathology is evident late in gestation as dilatations of the pancreatic ducts that continue to expand postnatally. Shortly after birth, the acini become disorganized, undergo apoptosis, and are largely ablated in late stage pathology. In addition, serum amylase levels are elevated and carboxypeptidase is abnormally activated within the pancreas. Ultrastructural analysis reveals that the acini undergo extensive vacuolization and have numerous 'halo-granules' similar to that seen in induced models of pancreatitis resulting from duct obstruction. Intriguingly, although the acini are severely affected in Tg737(orpk) mutants, cilia and Tg737 expression are restricted to the ducts and islets and are not detected on acinar cells. Analysis of the endocrine pancreas in Tg737(orpk) mutants revealed normal differentiation and distribution of cell types in the islets. However, after fasting, mutant blood glucose levels are significantly lower than controls and when challenged in glucose tolerance tests, Tg737(orpk) mutants exhibited defects in glucose uptake. These findings are interesting in light of the recently proposed role for polaris, the protein encoded by the Tg737 gene, in the hedgehog pathway and hedgehog signaling in insulin production and glucose homeostasis.


Assuntos
Pâncreas/anormalidades , Pâncreas/patologia , Proteínas Supressoras de Tumor , Animais , Animais Recém-Nascidos , Apoptose , Proliferação de Células , Cílios/ultraestrutura , Modelos Animais de Doenças , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Teste de Tolerância a Glucose , Ilhotas Pancreáticas/anormalidades , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Pâncreas/enzimologia , Pâncreas Exócrino/anormalidades , Pâncreas Exócrino/metabolismo , Pâncreas Exócrino/patologia , Ductos Pancreáticos/anormalidades , Ductos Pancreáticos/metabolismo , Ductos Pancreáticos/patologia , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , beta-Galactosidase/metabolismo
16.
J Physiol ; 547(Pt 1): 11-20, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12562919

RESUMO

There is ample evidence that an adverse intrauterine environment has harmful consequences for health in later life. Maternal diabetes and experimentally induced hyperglycaemia result in asymmetric overgrowth, which is associated with an increased insulin secretion and hyperplasia of the insulin-producing B-cells in the fetuses. In adult life, a reduced insulin secretion is found. In contrast, intrauterine growth restriction is associated with low insulin secretion and a delayed development of the insulin-producing B-cells. These perinatal alterations may induce a deficient adaptation of the endocrine pancreas and insulin resistance in later life. Intrauterine growth restriction in human pregnancy is mainly due to a reduced uteroplacental blood flow or to maternal undernutrition or malnutrition. However, intrauterine growth restriction can be present in severe diabetes complicated by vasculopathy and nephropathy. In animal models, intrauterine growth retardation can be obtained through pharmacological (streptozotocin), dietary (semi-starvation, low protein diet) or surgical (intrauterine artery ligation) manipulation of the maternal animal. The endocrine pancreas and more specifically the insulin-producing B-cells play an important role in the adaptation to an adverse intrauterine milieu and the consequences in later life. The long-term consequences of an unfavourable intrauterine environment are of major importance worldwide. Concerted efforts are needed to explore how these long-term effects can be prevented. This review will consist of two parts. In the first part, we discuss the long-term consequences in relation to the development of the fetal endocrine pancreas and fetal growth in the human; in the second part, we focus on animal models with disturbed fetal and pancreatic development and the consequences for later life.


Assuntos
Retardo do Crescimento Fetal/fisiopatologia , Ilhotas Pancreáticas/anormalidades , Ilhotas Pancreáticas/fisiopatologia , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Animais , Restrição Calórica , Feminino , Retardo do Crescimento Fetal/metabolismo , Humanos , Ilhotas Pancreáticas/metabolismo , Gravidez , Desnutrição Proteico-Calórica/metabolismo , Desnutrição Proteico-Calórica/fisiopatologia
17.
Trends Endocrinol Metab ; 13(5): 209-14, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12185667

RESUMO

Type 1 diabetes mellitus is a T-cell-mediated autoimmune disease that results in the destruction of the insulin-producing beta cells in the pancreatic islets of Langerhans. In spite of extensive genetic and immunological studies, mainly performed in the non-obese diabetic (NOD) spontaneous mouse model, the etiology of the autoimmune attack remains unknown. Several autoantigens have been identified and numerous studies have suggested a role for defective regulation of immune function. However, this account does not explain why the autoimmune process specifically affects the insulin-producing beta cells. Thus, abnormal immune regulation might explain the predisposition to autoimmunity in general, but additional factors should then determine the target of the autoimmune attack. Here, we review the evidence that abnormalities in islet cell differentiation and function exist that might trigger the immune system towards beta-cell autoimmunity in humans and NOD mice.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Animais Recém-Nascidos , Autoantígenos/genética , Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/genética , Feminino , Predisposição Genética para Doença , Humanos , Ilhotas Pancreáticas/anormalidades , Camundongos , Camundongos Endogâmicos NOD , Estado Pré-Diabético/genética , Estado Pré-Diabético/imunologia , Gravidez
18.
Am J Physiol Regul Integr Comp Physiol ; 283(3): R623-30, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12184996

RESUMO

Fetal malnutrition is now proposed as a risk factor of later obesity and type II diabetes. We previously analyzed the long-term impact of reduced protein and/or energy intake strictly limited to the last week of pregnancy in Wistar rats. Three protocols of gestational malnutrition were used: 1) low-protein isocaloric diet (5 instead of 15%) with pair feeding to the mothers receiving the control diet, 2) restricted diet (50% of control diet), and 3) low protein-restricted diet (50% of low-protein diet). Only isolated protein restriction induced a long-term beta-cell mass decrease. In the present study, we used the same protocols of food restriction to analyze their short-term impact (on day 21.5 of pregnancy) on beta-cell mass development. A 50% beta-cell mass decrease was present in the three restricted groups, but low-protein diet, either associated or not to energy restriction, increased fetal beta-cell insulin content. Among all the parameters analyzed to further explain our results, we found that the fetal plasma level of taurine was lowered by low-protein diet and was the main predictor of the fetal plasma insulin level (r = 0.63, P < 0.01). In conclusion, rat fetuses exposed to protein and/or energy restriction during the third part of pregnancy have a similar dramatic decrease in beta-cell mass, and their ability to recover beta-cell mass development retardation depends on the type of malnutrition used. Moreover, our results support the hypothesis that taurine might play an important role in fetal beta-cell mass function.


Assuntos
Ilhotas Pancreáticas/anormalidades , Insuficiência Placentária/patologia , Desnutrição Proteico-Calórica/patologia , Animais , Peso Corporal , Dieta com Restrição de Proteínas , Ingestão de Energia , Feminino , Idade Gestacional , Insulina/análise , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/patologia , Gravidez , Ratos , Ratos Wistar , Albumina Sérica , Taurina/sangue
19.
Int J Exp Diabetes Res ; 1(4): 289-98, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11467419

RESUMO

A failure in the compensate proliferation of pancreatic beta-cells, as the primary pathogenic event, has been reported in OLETF rat, a model of NIDDM. The aim of the present study is to define whether the beta-cell defect is attributed to the fetal stage islet development, if so, whether the defect involves down regulation of PDX-1 protein expression. Morphological changes, beta-cell function, and the expression of PDX-1 protein were examined in the cultured fetal islet-like cell clusters (ICCs) from OLETF rats along with their diabetes-resistant control counterpart LETO rats in the presence of 5.5 or 11.1mM glucose for 48, 72, 96, and 120-hr, respectively. We have observed four abnormalities in the ICCs of OLETF rats. First, a defective morphogenesis was noted during the 72 to 120-hr ICC culture, a period characterized by a dramatic increase in both beta-cell and non-beta-cell (alpha, delta, and PP) populations in control rats. This defective morphogenesis was demonstrated by a growth retardation of epithelial stratification and poor development of both beta-cell and non-beta-cell masses along with a parallel decline in relevant islet hormone contents. Second, a functional defect was characterized by failure to response to glucose during the 96 to 120-hr-cultured ICCs. Third, the ultrastructural analysis revealed a significant reduction in the number of secretory granules. Four, Western blot analysis showed a significant decrease of PDX-1 protein expression in the OLETF ICCs cultured in 11.1mM glucose for 48 to 72-hr and in 5.5mM glucose for 120-hr. Therefore, we concluded that during the fetal stage of islet development, OLETF rats exhibit both morphological and functional defects.


Assuntos
Diabetes Mellitus Tipo 2/embriologia , Proteínas de Homeodomínio , Ilhotas Pancreáticas/embriologia , Morfogênese , Animais , Glicemia/análise , Células Cultivadas , Feminino , Glucagon/análise , Insulina/análise , Insulina/sangue , Ilhotas Pancreáticas/anormalidades , Ilhotas Pancreáticas/química , Gravidez , Ratos , Ratos Endogâmicos OLETF , Fatores de Tempo , Transativadores/análise
20.
Nat Genet ; 23(1): 71-5, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10471502

RESUMO

In most mammals the pancreas develops from the foregut endoderm as ventral and dorsal buds. These buds fuse and develop into a complex organ composed of endocrine, exocrine and ductal components. This developmental process depends upon an integrated network of transcription factors. Gene targeting experiments have revealed critical roles for Pdx1, Isl1, Pax4, Pax6 and Nkx2-2 (refs 3,4,5,6,7, 8,9,10). The homeobox gene HLXB9 (encoding HB9) is prominently expressed in adult human pancreas, although its role in pancreas development and function is unknown. To facilitate its study, we isolated the mouse HLXB9 orthologue, Hlxb9. During mouse development, the dorsal and ventral pancreatic buds and mature beta-cells in the islets of Langerhans express Hlxb9. In mice homologous for a null mutation of Hlxb9, the dorsal lobe of the pancreas fails to develop. The remnant Hlxb9-/- pancreas has small islets of Langerhans with reduced numbers of insulin-producing beta-cells. Hlxb9-/- beta-cells express low levels of the glucose transporter Glut2 and homeodomain factor Nkx 6-1. Thus, Hlxb9 is key to normal pancreas development and function.


Assuntos
Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/anormalidades , Proteínas do Tecido Nervoso , Pâncreas/anormalidades , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho , Fatores de Transcrição Forkhead , Genótipo , Glucagon/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Insulina/metabolismo , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Proteínas com Homeodomínio LIM , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Neurônios Motores/metabolismo , Proteínas Nucleares , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Pâncreas/embriologia , Pâncreas/metabolismo , Polipeptídeo Pancreático/metabolismo , Proteínas Repressoras , Somatostatina/metabolismo , Fatores de Tempo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...