Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Front Immunol ; 12: 645817, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841427

RESUMO

T-cell responses to insulin and its precursor proinsulin are central to islet autoimmunity in humans and non-obese diabetic (NOD) mice that spontaneously develop autoimmune diabetes. Mice have two proinsulin genes proinsulin -1 and 2 that are differentially expressed, with predominant proinsulin-2 expression in the thymus and proinsulin-1 in islet beta-cells. In contrast to proinsulin-2, proinsulin-1 knockout NOD mice are protected from autoimmune diabetes. This indicates that proinsulin-1 epitopes in beta-cells maybe preferentially targeted by autoreactive T cells. To study the contribution of proinsulin-1 reactive T cells in autoimmune diabetes, we generated transgenic NOD mice with tetracycline-regulated expression of proinsulin-1 in antigen presenting cells (TIP-1 mice) with an aim to induce immune tolerance. TIP-1 mice displayed a significantly reduced incidence of spontaneous diabetes, which was associated with reduced severity of insulitis and insulin autoantibody development. Antigen experienced proinsulin specific T cells were significantly reduced in in TIP-1 mice indicating immune tolerance. Moreover, T cells from TIP-1 mice expressing proinsulin-1 transferred diabetes at a significantly reduced frequency. However, proinsulin-1 expression in APCs had minimal impact on the immune responses to the downstream antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) and did not prevent diabetes in NOD 8.3 mice with a pre-existing repertoire of IGRP reactive T cells. Thus, boosting immune tolerance to proinsulin-1 partially prevents islet-autoimmunity. This study further extends the previously established role of proinsulin-1 epitopes in autoimmune diabetes in NOD mice.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Tolerância Imunológica , Proinsulina/fisiologia , Animais , Autoanticorpos/análise , Glucose-6-Fosfatase/fisiologia , Insulina/imunologia , Camundongos , Camundongos Endogâmicos NOD , Proinsulina/genética , Proinsulina/imunologia
2.
Diabetes Obes Metab ; 20 Suppl 2: 95-103, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230182

RESUMO

ER stress due to proinsulin misfolding has an important role in the pathophysiology of rare forms of permanent neonatal diabetes (PNDM) and probably also of common type 1 (T1D) and type 2 diabetes (T2D). Accumulation of misfolded proinsulin in the ER stimulates the unfolded protein response (UPR) that may eventually lead to apoptosis through a process called the terminal UPR. However, the ß-cell ER has an incredible ability to cope with accumulation of misfolded proteins; therefore, it is not clear whether in common forms of diabetes the accumulation of misfolded proinsulin exceeds the point of no return in which terminal UPR is activated. Many studies showed that the UPR is altered in both T1D and T2D; however, the observed changes in the expression of different UPR markers are inconsistent and it is not clear whether they reflect an adaptive response to stress or indeed mediate the ß-cell dysfunction of diabetes. Herein, we critically review the literature on the effects of proinsulin misfolding and ER stress on ß-cell dysfunction and loss in diabetes with emphasis on ß-cell dynamics, and discuss the gaps in understanding the role of proinsulin misfolding in the pathophysiology of diabetes.


Assuntos
Diferenciação Celular , Diabetes Mellitus/etiologia , Células Secretoras de Insulina/fisiologia , Proinsulina/fisiologia , Dobramento de Proteína , Adaptação Fisiológica/fisiologia , Animais , Diferenciação Celular/fisiologia , Diabetes Mellitus/fisiopatologia , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/fisiologia , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Proinsulina/química , Suínos
4.
Trends Endocrinol Metab ; 21(11): 652-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20724178

RESUMO

Type 1B diabetes (typically with early onset and without islet autoantibodies) has been described in patients bearing small coding sequence mutations in the INS gene. Not all mutations in the INS gene cause the autosomal dominant Mutant INS-gene Induced Diabetes of Youth (MIDY) syndrome, but most missense mutations affecting proinsulin folding produce MIDY. MIDY patients are heterozygotes, with the expressed mutant proinsulins exerting dominant-negative (toxic gain of function) behavior in pancreatic beta cells. Here we focus primarily on proinsulin folding in the endoplasmic reticulum, providing insight into perturbations of this folding pathway in MIDY. Accumulated evidence indicates that, in the molecular pathogenesis of the disease, misfolded proinsulin exerts dominant effects that initially inhibit insulin production, progressing to beta cell demise with diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Proinsulina/química , Proinsulina/genética , Dobramento de Proteína , Sequência de Aminoácidos , Animais , Diabetes Mellitus Tipo 1/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Modelos Biológicos , Dados de Sequência Molecular , Mutação/fisiologia , Proinsulina/metabolismo , Proinsulina/fisiologia , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo
5.
Diabetes ; 59(8): 1937-47, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20530744

RESUMO

OBJECTIVE: Loss-of-function mutations in Perk (EIF2AK3) result in permanent neonatal diabetes in humans (Wolcott-Rallison Syndrome) and mice. Previously, we found that diabetes associated with Perk deficiency resulted from insufficient proliferation of beta-cells and from defects in insulin secretion. A substantial fraction of PERK-deficient beta-cells display a highly abnormal cellular phenotype characterized by grossly distended endoplasmic reticulum (ER) and retention of proinsulin. We investigated over synthesis, lack of ER-associated degradation (ERAD), and defects in ER to Golgi trafficking as possible causes. RESEARCH DESIGN AND METHODS: ER functions of PERK were investigated in cell culture and mice in which Perk was impaired or gene dosage modulated. The Ins2(+/Akita) mutant mice were used as a model system to test the role of PERK in ERAD. RESULTS: We report that loss of Perk function does not lead to uncontrolled protein synthesis but impaired ER-to-Golgi anterograde trafficking, retrotranslocation from the ER to the cytoplasm, and proteasomal degradation. PERK was also shown to be required to maintain the integrity of the ER and Golgi and processing of ATF6. Moreover, decreasing Perk dosage surprisingly ameliorates the progression of the Akita mutants toward diabetes. CONCLUSIONS: PERK is a positive regulator of ERAD and proteasomal activity. Reducing PERK activity ameliorates the progression of diabetes in the Akita mouse, whereas increasing PERK dosage hastens its progression. We speculate that PERK acts as a metabolic sensor in the insulin-secreting beta-cells to modulate the trafficking and quality control of proinsulin in the ER relative to the physiological demands for circulating insulin.


Assuntos
Proinsulina/fisiologia , eIF-2 Quinase/genética , eIF-2 Quinase/fisiologia , Animais , Clonagem Molecular , Diabetes Mellitus/genética , Retículo Endoplasmático/fisiologia , Dosagem de Genes , Glucose/farmacologia , Complexo de Golgi/fisiologia , Humanos , Recém-Nascido , Insulina/metabolismo , Secreção de Insulina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proinsulina/genética , Proinsulina/metabolismo , Transfecção , Translocação Genética , eIF-2 Quinase/deficiência
6.
Diabetologia ; 51(4): 597-601, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18264689

RESUMO

AIMS/HYPOTHESIS: Variation within six novel genetic loci has been reported to confer risk of type 2 diabetes and may be associated with beta cell dysfunction. We investigated whether these polymorphisms are also associated with impaired proinsulin to insulin conversion. METHODS: We genotyped 1,065 German participants for single nucleotide polymorphisms rs7903146 in TCF7L2, rs7754840 in CDKAL1, rs7923837 and rs1111875 in HHEX, rs13266634 in SLC30A8, rs10811661 in CDKN2A/B and rs4402960 in IGF2BP2. All participants underwent an OGTT. Insulin, proinsulin and C-peptide concentrations were measured at 0, 30, 60, 90 and 120 min during the OGTT. Insulin secretion was estimated from C-peptide or insulin levels during the OGTT using validated indices. We used the ratio proinsulin/insulin during the OGTT as indicator of proinsulin conversion. RESULTS: In our cohort, we confirmed the significant association of variants in TCF7L2, CDKAL1 and HHEX with reduced insulin secretion during the OGTT (p<0.05 for all). Variation in SLC30A8, CDKN2A/B and IGF2BP2 was not associated with insulin secretion. The risk alleles of the variants in TCF7L2, CDKAL1 and SLC30A8 reduced proinsulin to insulin conversion (p<0.05 for all), whereas the risk alleles in HHEX, CDKN2A/B and IGF2BP2 were not associated with reduced proinsulin to insulin conversion (p>0.6). CONCLUSIONS/INTERPRETATION: Diabetes-associated variants in TCF7L2 and CDKAL1 impair insulin secretion and conversion of proinsulin to insulin. However, both aspects of beta cell function are not necessarily linked, as impaired insulin secretion is specifically present in variants of HHEX and impaired proinsulin conversion is specifically present in a variant of SLC30A8.


Assuntos
Proteínas de Transporte de Cátions/genética , Quinase 5 Dependente de Ciclina/genética , Diabetes Mellitus/genética , Insulina/metabolismo , Polimorfismo de Nucleotídeo Único , Proinsulina/fisiologia , Fatores de Transcrição TCF/genética , Adulto , Área Sob a Curva , Peptídeo C/sangue , Feminino , Alemanha , Proteínas de Homeodomínio/genética , Humanos , Secreção de Insulina , Masculino , Pessoa de Meia-Idade , Proinsulina/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição , Fatores de Transcrição/genética , Transportador 8 de Zinco , tRNA Metiltransferases
7.
Rom J Intern Med ; 45(1): 3-15, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17966438

RESUMO

The main beta cell function is that of pre-proinsulin synthesis and of insulin exocytosis in a regulated manner. After the detachment of a small signal peptide, the remaining proinsulin molecule is transferred in the endoplasmic reticulum. During the emergence of the secretory vesicles and their maturation, proinsulin is split into insulin and C peptide. Diabetes mellitus is characterized by a poor maturation of secretory vesicles explaining the higher levels of proinsulin both in beta cells and in the plasma. The defect is associated to alterations in the exocytosis machinery, initially minor (disappearance of oscillatory pattern of insulin release, and/or the amputation of early phase of insulin response) and later major (a progressive decrease of the overall insulin response). Because the increase in plasma glucose levels is a late indicator of the diabetogenic process (a decrease with more than 50% of beta cell mass/function), we propose as marker of the prehyperglycaemia the high levels of plasma proinsulin or of the proinsulin/insulin ratio. In type 1 diabetes, the autoimmune destruction of beta cell mass will have a fast evolution, while in the type 2 phenotype, the same process takes a slower, but also progressive evolution. In both cases, the decrease in beta cell mass will be induced by an increased apoptosis and the decreased regeneration reaction.


Assuntos
Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Humanos , Células Secretoras de Insulina/fisiologia , Proinsulina/fisiologia , Vesículas Secretórias/fisiologia
8.
Diabetologia ; 49(6): 1142-50, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16596360

RESUMO

In postnatal organisms, insulin is well known as an essential anabolic hormone responsible for maintaining glucose homeostasis. Its biosynthesis by the pancreatic beta cell has been considered a model of tissue-specific gene expression. However, proinsulin mRNA and protein have been found in embryonic stages before the formation of the pancreatic primordium, and later, in extrapancreatic tissues including the nervous system. Phylogenetic studies have also confirmed that production of insulin-like peptides antecedes the morphogenesis of a pancreas, and that these peptides contribute to normal development. In recent years, other roles for insulin distinct from its metabolic function have emerged also in vertebrates. During embryonic development, insulin acts as a survival factor and is involved in early morphogenesis. These findings are consistent with the observation that, at these stages, the proinsulin gene product remains as the precursor form, proinsulin. Independent of its low metabolic activity, proinsulin stimulates proliferation in developing neuroretina, as well as cell survival and cardiogenesis in early embryos. Insulin/proinsulin levels are finely regulated during development, since an excess of the protein interferes with correct morphogenesis and is deleterious for the embryo. This fine-tuned regulation is achieved by the expression of alternative embryonic proinsulin transcripts that have diminished translational activity.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Proinsulina/fisiologia , Envelhecimento , Animais , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Ilhotas Pancreáticas/embriologia , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Filogenia , Proinsulina/genética
10.
Clin Lab ; 50(9-10): 567-73, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15481632

RESUMO

Proinsulin, the precursor of insulin during physiological insulin production, has been demonstrated in the past to stimulate PAI-1 secretion and consecutively block fibrinolysis. Therefore, proinsulin is contributing as an independent factor to the increased cardiovascular risk of patients with type 2 diabetes. However, development of insulin resistance in the course of type 2 diabetes leads to increased insulin demands and finally to an impairment of beta-cell function in later disease stages. Appearance of intact proinsulin in the peripheral blood has been shown to be a good laboratory marker for this phenomenon since it indicates an exhaustion of the cleavage capacity of the intracellular processing enzymes. However, the close relation of the two pathophysiological entities also makes it a very specific marker for insulin resistance per se. During the past years, new immunoassays have been developed that are able to distinguish between intact proinsulin and its specific and unspecific cleavage products. Use of these assays in recent epidemiological and intervention studies has helped to get a better understanding about beta-cell dysfunction and its relation to insulin resistance and cardiovascular risk. In a large cross-sectional study with 4270 orally treated patients, elevation of fasting intact proinsulin was very closely related to insulin resistance, as assessed by iv glucose tolerance test in a subgroup, and by HOMA analysis in the entire patient population. Effective treatment of insulin resistance (e.g. with thiazolidindiones) led to a decrease in elevated proinsulin levels and to a decrease of the cardiovascular risk profile, while the levels remained high during sulfonylurea therapy. These results suggest to reconsider intact and total proinsulin as valuable diagnostic tools in diagnosis and treatment of type 2 diabetes. Based on the published data of the new specific immunoassays, patients with elevated intact proinsulin levels (> 10 pmol/L) should be regarded and treated as being insulin-resistant, while elevation of total proinsulin (>45 pmol/l) may help to identify the high cardiovascular risk patients. Both assays can thus be used to assess beta-cell function, to facilitate the selection of the most promising therapy, and may also serve to monitor treatment success in the further course of the disease.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Proinsulina/fisiologia , Biomarcadores/sangue , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/fisiopatologia , Humanos , Imunoensaio , Proinsulina/sangue
11.
Exp Diabesity Res ; 5(1): 7-14, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15198367

RESUMO

The C-peptide links the insulin A and B chains in proinsulin, providing thereby a means to promote their efficient folding and assembly in the endoplasmic reticulum during insulin biosynthesis. It then facilitates the intracellular transport, sorting, and proteolytic processing of proinsulin into biologically active insulin in the maturing secretory granules of the beta cells. These manifold functions impose significant constraints on the C-peptide structure that are conserved in evolution. After cleavage of proinsulin, the intact C-peptide is stored with insulin in the soluble phase of the secretory granules and is subsequently released in equimolar amounts with insulin, providing a useful independent indicator of insulin secretion. This brief review highlights many aspects of its roles in biosynthesis, as a prelude to consideration of its possible additional role(s) as a physiologically active peptide after its release with insulin into the circulation in vivo.


Assuntos
Peptídeo C/fisiologia , Proinsulina/fisiologia , Sequência de Aminoácidos , Animais , Peptídeo C/genética , Evolução Molecular , Humanos , Insulina/biossíntese , Conformação Molecular , Dados de Sequência Molecular , Proinsulina/genética , Processamento de Proteína Pós-Traducional
12.
Pediatrics ; 113(3 Pt 1): 537-41, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14993546

RESUMO

OBJECTIVE: Many extremely preterm infants develop hyperglycemia in the first week of life during continuous glucose infusion. The objective of this study was to determine whether defective insulin secretion or resistance to insulin was the primary factor involved in transient hyperglycemia of extremely preterm infants. METHODS: A prospective comparative study was conducted in appropriate-for-gestational-age preterm infants <30 weeks of gestational age with the aim specifically to evaluate the serum levels of proinsulin, insulin, and C-peptide secreted during transient hyperglycemia by specific immunoassays. Three groups of infants were investigated hyperglycemic (n = 15) and normoglycemic preterm neonates (n = 12) and normal, term neonates (n = 21). In addition, the changes in beta-cell peptide levels were analyzed during and after intravenous insulin infusion in the hyperglycemic group. Data were analyzed using analysis of variance and analysis of variance for repeated measures. RESULTS: At inclusion, insulin and C-peptide levels did not differ in hyperglycemic subjects and in preterm controls. Proinsulin concentration was significantly higher in the hyperglycemic group (36.5 +/- 3.9 vs 23.2 +/- 0.9 pmol/L). Compared with term neonates, proinsulin and C-peptide levels were higher in normoglycemic preterm infants (23.2 +/- 0.9 vs 18.9 +/- 2.71 pmol/L and 1.67 +/- 0.3 vs 0.62 +/- 0.12 nmol/L, respectively). During and after insulin infusion in hyperglycemic neonates, plasma glucose concentration fell and proinsulin and C-peptide levels were lowered (18.4 +/- 7.6 and 20.7 +/- 4.5 pmol/L, respectively). CONCLUSION: These data suggest that 1) preterm neonates are sensitive to changes in plasma glucose concentration, but proinsulin processing to insulin is partially defective in hyperglycemic preterm neonates; 2) hyperglycemic neonates are relatively resistant to insulin because higher insulin levels are needed to achieve euglycemia in this group compared with normoglycemic neonates. These results also show that insulin infusion is beneficial in extremely preterm infants with transient hyperglycemia.


Assuntos
Hiperglicemia/sangue , Doenças do Prematuro/sangue , Resistência à Insulina/fisiologia , Proinsulina/fisiologia , Glicemia , Peptídeo C/sangue , Feminino , Humanos , Hiperglicemia/etiologia , Recém-Nascido , Recém-Nascido Prematuro/fisiologia , Doenças do Prematuro/etiologia , Insulina/administração & dosagem , Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Masculino , Proinsulina/sangue , Estudos Prospectivos
13.
Diabetes Metab Res Rev ; 19(5): 345-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12951641

RESUMO

Proinsulin C-peptide was for long considered to be without biological activity of its own. New findings demonstrate, however, that it is capable of eliciting both molecular and physiological effects, suggesting that C-peptide is in fact a bioactive peptide. When administered in replacement doses to animal models or to patients with type 1 diabetes, C-peptide ameliorates diabetes-induced functional and structural changes in both the kidneys and the peripheral nerves. It augments blood flow in a number of tissues, notably skeletal muscle, myocardium, skin and nerve. These effects are thought to be mediated via a stimulatory influence on Na+,K(+)-ATPase and on endothelial nitric oxide synthase. Specific binding of C-peptide to cell membranes of intact cells and to detergent-solubilized cellular components has been demonstrated, indicating the existence of cell-surface binding sites for C-peptide. A number of intracellular responses are elicited by C-peptide, including a rise in Ca2+ concentration and activation of MAP-kinase signaling pathways. Many but not all of C-peptide's intracellular effects can be inhibited by pertussis toxin, supporting the notion that C-peptide may interact via a G-protein-coupled receptor. Additional data suggest that C-peptide may interact synergistically also in the insulin signaling pathway. Combined, the available observations show conclusively that C-peptide is biologically active, even though its molecular mechanism of action is not as yet fully understood. The possibility that replacement of C-peptide in patients with type 1 diabetes may serve to retard or prevent the development of long-term complications should be evaluated.


Assuntos
Peptídeo C/fisiologia , Peptídeo C/genética , Peptídeo C/uso terapêutico , Sequência Conservada , Diabetes Mellitus Tipo 1/tratamento farmacológico , Humanos , Proinsulina/fisiologia
14.
Proc Natl Acad Sci U S A ; 100(18): 10376-81, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12925730

RESUMO

It has been reported that an insulin 2 gene knockout, when bred onto nonobese diabetic (NOD) mice, accelerates diabetes. We produced insulin 1 gene knockout congenic NOD mice. In contrast to insulin 2, diabetes and insulitis were markedly reduced in insulin 1 knockout mice, with decreased and delayed diabetes in heterozygous females and no insulitis and diabetes in most homozygous female mice. Lack of insulitis was found for insulin 1 female homozygous knockout mice at 8, 12, and 37 weeks of age. Despite a lack of insulitis, insulin 1 homozygous knockout mice spontaneously expressed insulin autoantibodies. Administration of insulin peptide B:9-23 of both insulin 1 and 2 to NOD mice induced insulin autoantibodies. Insulin 1 is not the only lymphocytic target of NOD mice. Insulin 1 homozygous knockout islets, when transplanted into recently diabetic wild-type NOD mice, became infiltrated with lymphocytes and only transiently reversed diabetes. These observations indicate that loss of either insulin gene can influence progression to diabetes of NOD mice and suggest that the preproinsulin 1 gene is crucial for the spontaneous development of NOD insulitis and diabetes.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Ilhotas Pancreáticas/patologia , Proinsulina/fisiologia , Precursores de Proteínas/fisiologia , Animais , Feminino , Insulina , Anticorpos Anti-Insulina/sangue , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Proinsulina/genética , Precursores de Proteínas/genética , Linfócitos T/imunologia
15.
J Clin Invest ; 111(6): 851-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12639991

RESUMO

Accumulating evidence favors a role for proinsulin as a key autoantigen in diabetes. In the mouse, two proinsulin isoforms coexist. Most studies point to proinsulin 2 as the major isoform recognized by T cells in the NOD mouse. We studied mice in which a null proinsulin 2 mutation was transferred from proinsulin 2-deficient 129 mice onto the NOD background along with 16 genetic markers (including I-A(g7) MHC molecule) associated with diabetes. Intercross mice from the fourth backcross generation showed that proinsulin 2(-/-) mice develop accelerated insulitis and diabetes. The high prevalence of anti-insulin autoantibodies in proinsulin 2(-/-) mice indicates that diabetes acceleration relates to altered recognition of proinsulin. The prevalence of anti-glutamic acid decarboxylase autoantibodies and of sialitis is not increased in proinsulin 2(-/-) mice. We give evidence that proinsulin 2 expression leads to silencing of T cells specific for an epitope shared by proinsulin 1 and proinsulin 2. In the human, alleles located in the VNTR region flanking the insulin gene control beta cell response to glucose and proinsulin expression in the thymus and are key determinants of diabetes susceptibility. Proinsulin 2(-/-) NOD mice provide a model to study the role of thymic expression of insulin in susceptibility to diabetes.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Proinsulina/fisiologia , Transferência Adotiva , Sequência de Aminoácidos , Animais , Autoimunidade , Diabetes Mellitus Tipo 1/imunologia , Glutamato Descarboxilase/imunologia , Anticorpos Anti-Insulina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Dados de Sequência Molecular , Proinsulina/deficiência
16.
Diabetes ; 51 Suppl 3: S489-93, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12475795

RESUMO

Insulin is a major disease determinant in type 1 diabetes, type 2 diabetes, and related disorders. The role of variations in the expression of the insulin gene has been proposed in genetic susceptibility to the three pathological conditions in humans. In contrast to humans, rodents express two proinsulin isoforms. One isoform, proinsulin 1, is expressed exclusively in islets. The second, proinsulin 2, is expressed in islets and in other tissues, especially the thymus. We took advantage of the expression of these two isoforms to introduce a null proinsulin 2 allele in NOD mice and to evaluate the consequence of a variation of proinsulin 2 gene expression on the development of type 1 diabetes on the NOD genetic background. Heterozygote NOD mutant mice carrying a null proinsulin 2 mutation showed an increased incidence of type 1 diabetes at successive backcross generations. Plasma glucose and insulin levels were identical in prediabetic mutant and in wild-type mice at 4 weeks of age. Variation in insulin gene expression is hypothesized to interfere with diabetes development at both the islet and the thymus level.


Assuntos
Diabetes Mellitus Tipo 1/genética , Expressão Gênica , Variação Genética , Insulina/genética , Camundongos Endogâmicos NOD/fisiologia , Proinsulina/fisiologia , Animais , Predisposição Genética para Doença/genética , Camundongos , Camundongos Endogâmicos NOD/genética , Camundongos Knockout/genética , Proinsulina/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia
17.
Int J Exp Diabetes Res ; 2(2): 145-51, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12369718

RESUMO

In recent years the physiological role of the proinsulin C-peptide has received increasing attention, focusing on the potential therapeutic value of C-peptide replacement in preventing and ameliorating type 1 diabetic complications. In order to consolidate these new data and to identify the immediate directions of C-peptide research and its clinical usefulness, an International Symposium was held in Detroit, Michigan, on October 20-21, 2000, under the auspices of the Wayne State University/Morris Hood Jr. Comprehensive Diabetes Center. In this communication, we review the cellular, physiological and clinical effects of C-peptide replacement in animal models and in patients with type 1 diabetes. Finally, recommendations are presented as to the most urgent studies that should be pursued to further establish the biological action of C-peptide and its therapeutic value.


Assuntos
Peptídeo C/fisiologia , Diabetes Mellitus/fisiopatologia , Proinsulina/fisiologia , Apoptose , Ensaios Clínicos como Assunto , Diabetes Mellitus/sangue , Humanos , ATPase Trocadora de Sódio-Potássio/metabolismo
19.
Gene Ther ; 7(3): 205-14, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10694797

RESUMO

Insulin gene therapy requires that insulin secretion be coupled to metabolic requirements. To this end, we have developed an insulin transgene whose transcription is stimulated by glucose and inhibited by insulin. Glucose- and insulin-sensitive promoters were constructed by inserting glucose-responsive elements (GlREs) from the rat L-pyruvate kinase (L-PK) gene into the insulin-sensitive, liver-specific, rat insulin-like growth factor binding protein-1 (IGFBP-1) promoter. Glucose (5 to 25 mM) stimulated, and insulin (10-10 to 10-7 M) inhibited, luciferase expression driven by these promoters in primary cultured rat hepatocytes. The capacity of transfected hepatocytes to secrete mature, biologically active insulin was demonstrated using a human proinsulin cDNA (2xfur), modified to allow protein processing by endogenous endopeptidase activity. Medium conditioned by insulin-producing hepatocytes contained greater than 300 microU/ml immunoreactive insulin, while denaturing SDS-PAGE of an anti-insulin immunoprecipitate revealed bands with the mobilities of insulin A, and B chains. Biological activity of hepatocyte-produced insulin was demonstrated in a transfection assay, in which medium conditioned by insulin-producing hepatocytes exerted an effect similar to 10-7 M insulin. We then combined the glucose- and insulin-sensitive promoter with the modified human proinsulin cDNA to create a metabolically sensitive insulin transgene ((GlRE)3BP-1 2xfur). In both H4IIE hepatoma cells stably transfected with this construct, and normal rat hepatocytes (GlRE)3BP-1 2xfur-mediated insulin secretion increased in response to stimulation by glucose. Moreover, a capacity to decrease insulin production in response to diminishing glucose exposure was also demonstrated. We conclude that the transcriptional regulation of insulin production using these glucose- and insulin-sensitive constructs meets the requirements for application in a rodent model of insulin gene therapy. Gene Therapy (2000) 7, 205-214.


Assuntos
Glucose/fisiologia , Insulina/genética , Transgenes/genética , Animais , Diabetes Mellitus/patologia , Diabetes Mellitus/terapia , Expressão Gênica , Humanos , Insulina/metabolismo , Fígado/metabolismo , Fígado/patologia , Proinsulina/fisiologia , Regiões Promotoras Genéticas/genética , Ratos
20.
Diabetes Care ; 22(4): 609-14, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10189540

RESUMO

OBJECTIVE: Type 2 diabetes is a heterogeneous disease in which both beta-cell dysfunction and insulin resistance are pathogenetic factors. Disproportionate hyperproinsulinemia (elevated proinsulin/insulin) is another abnormality in type 2 diabetes whose mechanism is unknown. Increased demand due to obesity and/or insulin resistance may result in secretion of immature beta-cell granules with a higher content of intact proinsulin. RESEARCH DESIGN AND METHODS: We investigated the impact of obesity on beta-cell secretion in normal subjects and in type 2 diabetic patients by measuring intact proinsulin, total proinsulin immunoreactivity (PIM), intact insulin, and C-peptide (by radioimmunoassay) by specific enzyme-linked immunosorbent assays in the fasting state and during a 120-min glucagon (1 mg i.v.) stimulation test. Lean (BMI 23.5 +/- 0.3 kg/m2) (LD) and obese (30.1 +/- 0.4 kg/m2) (OD) type 2 diabetic patients matched for fasting glucose (10.2 +/- 0.6 vs. 10.3 +/- 0.4 mmol/l) were compared with age- and BMI-matched lean (22.4 +/- 0.6 kg/m2) (LC) and obese (30.8 +/- 0.9 kg/m2) (OC) normal control subjects. RESULTS: Diabetic patients (LD vs. LC and OD vs. OC) had elevated fasting levels of intact proinsulin 6.6 +/- 1.0 vs. 1.6 +/- 0.3 pmol/l and 7.7 +/- 2.0 vs. 1.2 +/- 0.2 pmol/l; PIM: 19.9 +/- 2.5 vs. 5.4 +/- 1.0 pmol/l and 29.6 +/- 6.1 vs. 6.1 +/- 0.9 pmol/l; and total PIM/intact insulin: 39 +/- 4 vs. 15 +/- 2% and 35 +/- 5 vs. 13 +/- 2%, all P < 0.01. After glucagon stimulation, PIM levels were disproportionately elevated (PIM/intact insulin based on area under the curve analysis) in diabetic patients (LD vs. LC and OD vs. OC): 32.6 +/- 6.7 vs. 9.2 +/- 1.1% and 22.7 +/- 5.2 vs. 9.1 +/- 1.1%, both P < 0.05. Intact insulin and C-peptide net responses were significantly reduced in type 2 diabetic patients, most pronounced in the lean group. The ratio of intact proinsulin to PIM was higher in diabetic patients after stimulation in both LD versus LC: 32 +/- 3 vs. 23 +/- 2%, and OD versus OC: 28 +/- 4 vs. 16 +/- 2%, both P < 0.01. In obese normal subjects, intact proinsulin/PIM was lower both in the fasting state and after glucagon stimulation: OC versus LC: 22 +/- 3 vs. 33 +/- 3% (fasting) and 16 +/- 2 vs. 23 +/- 2% (stimulated), both P < 0.05. CONCLUSIONS: Increased secretory demand from obesity-associated insulin resistance cannot explain elevated intact proinsulin and disproportionate hyperproinsulinemia in type 2 diabetes. This abnormality may be an integrated part of pancreatic beta-cell dysfunction in this disease.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Ilhotas Pancreáticas/fisiologia , Obesidade/fisiopatologia , Proinsulina/fisiologia , Índice de Massa Corporal , Peptídeo C/análise , Diabetes Mellitus/fisiopatologia , Ensaio de Imunoadsorção Enzimática , Jejum , Feminino , Glucagon , Humanos , Masculino , Pessoa de Meia-Idade , Radioimunoensaio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...