Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nutr Diabetes ; 7(12): 305, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29269872

RESUMO

Free fatty acids (FFAs) can cause glucose intolerance and diabetes. Lipotoxicity to the pancreatic beta cells is considered to be a major underlying cause for this phenomenon. The aim of this study was to analyse the toxicity profile of FFAs in the human EndoC-ßH1 beta-cell line and to compare the results with isolated rat and human islets with special reference to the physiologically most prevalent FFAs palmitic acid (PA) and oleic acid (OA). Toxicity after a 2-day incubation with the different FFAs was analysed by the caspase-3 assay and confirmed by the propidium iodide and annexin V staining tests. The long-chain saturated PA (C16:0) and the monounsaturated OA (C18:1) were both toxic to human EndoC-ßH1 beta cells and pseudoislets, as well as to rat islets, and, as confirmed in a pilot experiment, also to human islets. Furthermore, OA provided no protection against the toxicity of PA. Likewise, elaidic acid (EA, the trans isomer of OA; trans-OA) was significantly toxic, in contrast to the non-metabolisable analogues methylated PA (MePA) and methylated OA (MeOA). Fatty acids with a chain length < C16 were not toxic in EndoC-ßH1 beta cells. Caspase-3 was also activated by linoleic acid (LA)(C18:2) but not by γ-linolenic acid (γ-LNA)(C18:3). Overall, only long-chain FFAs with chain lengths > C14, which generate hydrogen peroxide in the peroxisomal beta-oxidation, were toxic. This conclusion is also supported by the toxicity of the branched-chain FFA pristanic acid, which is exclusively metabolised in the peroxisomal beta-oxidation. The lack of a protective effect of the monounsaturated fatty acid OA has important consequences for a beta-cell protective lipid composition of a diet. A cardioprotective diet with a high OA content does not fulfil this requirement.


Assuntos
Ácidos Graxos Monoinsaturados/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Ácido Oleico/toxicidade , Ácido Palmítico/toxicidade , Animais , Caspase 3/metabolismo , Linhagem Celular , Humanos , Células Secretoras de Insulina/metabolismo , Ratos , Ratos Endogâmicos Lew
2.
Nutr Diabetes ; 4: e123, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-24979152

RESUMO

OBJECTIVE: Epidemiological evidence shows that chronic coffee consumption in humans is correlated with a lower incidence of type 2 diabetes mellitus. For the experimental exploration of the underlying mechanisms, this effect needs to be replicated in an animal model of type 2 diabetes with a short lifespan. DESIGN: Male C57BL/6 mice consumed regular coffee or water ad libitum and the development of obesity and diabetes caused by high-fat diet (55% lipids, HFD) was observed from week 10 on for 35 weeks in comparison with mice feeding on a defined normal diet (9% lipids, ND). RESULTS: The massive weight gain in HFD mice was dose-dependently retarded (P=0.034), the moderate weight gain in ND mice was abolished (P<0.001) by coffee consumption, probably because of a lower feeding efficiency. The consumption of fluid (water or coffee) was significantly diminished by HFD (P<0.001), resulting in a higher coffee exposure of ND mice. On week 21 intraperitoneal glucose tolerance tests (IPGTT) showed a dose-dependent faster decline of elevated glucose levels in coffee-consuming HFD mice (P=0.016), but not in ND mice. Remarkably, a spontaneous decrease in non-fasting glycaemia occurred after week 21 in all treatment groups (P<0.001). On week 39 the IPGTT showed diminished peak of glucose levels in coffee-consuming HFD mice (P<0.05). HFD mice were hyperinsulinaemic and had significantly (P<0.001) enlarged islets. Coffee consumption did not affect islet size or parameters of beta-cell apoptosis, proliferation and insulin granule content. CONCLUSION: Coffee consumption retarded weight gain and improved glucose tolerance in a mouse model of type 2 diabetes and corresponding controls. This gives rise to the expectation that further insight into the mechanism of the diabetes-preventive effect of coffee consumption in humans may be gained by this approach.

3.
Clin Exp Immunol ; 177(1): 168-78, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24628466

RESUMO

The LEW.1AR1-iddm rat is an animal model of human type 1 diabetes (T1D), which arose through a spontaneous mutation within the major histocompatibility complex (MHC)-congenic background strain LEW.1AR1. The LEW.1AR1-iddm rat is characterized by two phenotypes: diabetes development with a diabetes incidence of 60% and a variable T cell frequency in peripheral blood. In this study the immune cell repertoire of LEW.1AR1-iddm rats was analysed over time from days 30 to 90 of life and compared to the background strain LEW.1AR1 and the LEW rat strain as well as the LEW.1WR1 rat strain. The LEW.1AR1-iddm rats are characterized by a high variability of CD3(+), CD4(+) and CD8(+) T cell frequencies in peripheral blood over time, and the frequency is unique for each animal. The variability within the frequencies resulted in changes of the CD4(+) : CD8(+) T cell ratio. The other three rat strains studied were characterized by a stable but nevertheless strain-specific T cell frequency resulting in a specific CD4(+) : CD8(+) T cell ratio. The frequency of natural killer (NK) cells and B cells in LEW.1AR1-iddm rats was increased, with a higher variability compared to the other strains. Only monocytes showed no differences in frequency and variability between all strains studied. These variabilities of immune cell frequencies in the LEW.1AR1-iddm rats might lead to imbalances between autoreactive and regulatory T cells in peripheral blood as a prerequisite for diabetes development.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Células Matadoras Naturais/imunologia , Animais , Animais Congênicos , Circulação Sanguínea/imunologia , Relação CD4-CD8 , Diabetes Mellitus Tipo 1/genética , Modelos Animais de Doenças , Progressão da Doença , Predisposição Genética para Doença , Homeostase , Humanos , Ratos , Ratos Endogâmicos Lew , Ratos Mutantes , Fatores de Tempo
4.
Diabetologia ; 52(7): 1381-90, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19367386

RESUMO

AIMS/HYPOTHESIS: The LEW.1AR1-iddm rat is an animal model of spontaneous type 1 diabetes mellitus. This study analysed how adoptive transfer of selective T cell subpopulations affects the incidence of diabetes. METHODS: CD4(+) or CD8(+) T cells were isolated from diabetic LEW.1AR1-iddm rats or diabetes-resistant LEW.1AR1 rats. Cells were selectively transferred into athymic LEW.1AR1-Whn ( rnu ) or prediabetic LEW.1AR1-iddm rats. The animals were monitored for blood glucose, islet infiltration and immune cell composition of pancreas-draining lymph nodes. RESULTS: After adoptive transfer of CD4(+) T cells from diabetic LEW.1AR1-iddm rats into athymic LEW.1AR1-Whn ( rnu ) rats, 50% of the recipients developed diabetes. Transfer of CD8(+) T cells failed to induce diabetes. Only 10% of the athymic recipients became diabetic after co-transfer of CD4(+) and CD8(+) T cells. Adoptive transfer of CD8(+) T cells from LEW.1AR1 or diabetic LEW.1AR1-iddm rats into prediabetic LEW.1AR1-iddm rats significantly reduced the incidence of diabetes. In protected normoglycaemic animals regulatory CD8(+)/CD25(+) and CD4(+)/CD25(+) T cell subpopulations that were also FOXP3-positive accumulated in the pancreas-draining lymph nodes. In this lymphatic organ, gene expression of anti-inflammatory cytokines was significantly higher than in diabetic rats. CONCLUSIONS/INTERPRETATION: Our results show that adoptive transfer of CD4(+) but not CD8(+) T cells from diabetic LEW.1AR1-iddm rats induced diabetes development. Importantly, CD8(+) T cells from diabetic LEW.1AR1-iddm rats and diabetes-resistant LEW.1AR1 rats provided protection against beta cell destruction. The accumulation of regulatory T cells in the pancreas-draining lymph nodes from protected rats indicates that transferred CD8(+) T cells may have beneficial effects in the control of beta cell autoimmunity.


Assuntos
Transferência Adotiva/métodos , Linfócitos T CD8-Positivos/transplante , Diabetes Mellitus Tipo 1/prevenção & controle , Linfonodos/imunologia , Pâncreas/imunologia , Estado Pré-Diabético/terapia , Animais , Glicemia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/imunologia , Citocinas/genética , Citocinas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Expressão Gênica/imunologia , Imunofenotipagem , Estado Pré-Diabético/imunologia , Ratos , Ratos Endogâmicos Lew , Ratos Nus , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
5.
Cell Prolif ; 41(4): 607-24, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18616698

RESUMO

OBJECTIVES: Embryonic stem cells are a potential source for insulin-producing cells, but existing differentiation protocols are of limited efficiency. Here, the aim has been to develop a new one, which drives development of embryonic stem cells towards insulin-producing cells rather than to neuronal cell types, and to combine this with a strategy for their separation from insulin-negative cells. MATERIALS AND METHODS: The cytokeratin-19 (CK19) promoter was used to control the expression of enhanced yellow fluorescence protein in mouse embryonic stem cells during their differentiation towards insulin-producing cells, using a new optimized four-stage protocol. Two cell populations, CK19(+) and CK19(-) cells, were successfully fluorescence sorted and analysed. RESULTS: The new method reduced neuronal progeny and suppressed differentiation into glucagon- and somatostatin-producing cells. Concomitantly, beta-cell like characteristics of insulin-producing cells were strengthened, as documented by high gene expression of the Glut2 glucose transporter and the transcription factor Pdx1. This novel protocol was combined with a cell-sorting technique. Through the combined procedure, a fraction of glucose-responsive insulin-secreting CK19(+) cells was obtained with 40-fold higher insulin gene expression and 50-fold higher insulin content than CK19(-) cells. CK19(+) cells were immunoreactive for C-peptide and had ultrastructural characteristics of an insulin-secretory cell. CONCLUSION: Differentiated CK19(+) cells reflect an endocrine precursor cell type of ductal origin, potentially suitable for insulin replacement therapy in diabetes.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Insulina/genética , Queratina-19/biossíntese , Animais , Linhagem Celular , Separação Celular/métodos , Meios de Cultura , Primers do DNA , Células-Tronco Embrionárias/ultraestrutura , Citometria de Fluxo , Glucagon/genética , Camundongos , Plasmídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somatostatina/genética , Transfecção
7.
Diabetologia ; 49(11): 2704-12, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17047921

RESUMO

AIMS/HYPOTHESIS: It has recently been proposed that IL-1beta may be responsible for beta cell death in type 2 diabetes mellitus. Major support for this assumption was derived from experiments in the gerbil Psammomys obesus (sand rat), a model for nutritionally induced non-insulin-dependent type 2 diabetes. Using gerbil-specific primers for the analysis of gene expression, we investigated the validity of this hypothesis. METHODS: Gene expression was analysed by real-time RT-PCR of isolated and laser-microdissected islets and by in situ RT-PCR, both in beta cells and in immune cells, as well as in lymph nodes and spleen. RESULTS: We were unable to detect Il-1beta and the IL-1beta-inducible enzyme inducible nitric oxide synthase (iNos) by in situ RT-PCR, either in the pancreatic beta cells, or in the small number of non-activated immune cells of healthy and diabetic Psammomys obesus after 1 and 3 weeks on a high-energy diet. Very low levels of Il-1beta and iNos mRNA were detectable in collagenase-isolated and laser-microdissected islets of normoglycaemic gerbils by real-time RT-PCR without any increase of these mRNAs in islets from diabetic animals. These results were confirmed by electron microscopy with immunogold staining for IL-1beta and insulin. CONCLUSIONS/INTERPRETATION: The diabetic syndrome induced in Psammomys obesus by high-energy diet is a classical type 2 diabetes model, which does not show any evidence of an involvement of the proinflammatory cytokine IL-1beta or of activated immune cells in its pathogenesis. This is clearly at variance with the situation in type 1 diabetes.


Assuntos
Citocinas/fisiologia , Hiperglicemia/patologia , Células Secretoras de Insulina/patologia , Animais , Morte Celular , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Gerbillinae , Células Secretoras de Insulina/enzimologia , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Transplant Proc ; 35(8): 3147-52, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14698000

RESUMO

AIM: The pattern of rejection of vascularized pancreas allografts in presensitized recipients was studied among congeneic and recombinant rat strains in order to establish the time frame of rejection. Donors and recipients differed either in their entire MHC or in class I or class II MHC antigens. MATERIAL AND METHODS: Streptozotocin diabetic (55 mg/kg bone weight) recipients underwent an intra-abdominal heterotopic pancreas whole organ transplantation. The exocrine secretion of the pancreas was suppressed by ligation of the duct. Rejection was defined as recurrence of diabetic hyperglycemia exceeding 14 mmol/L, which was subsequently confirmed by histologic examination. The pancreas recipients were presensitized by repetitive donor-specific skin transplants performed 12, 8, and 4 weeks prior to pancreas grafting. RESULTS: The results shown accelerated rejection in all instances. Animals with incompatibility for the entire MHC showed a median rejection time in naive recipients of 12.5 days and in presensitized recipients of 8 days. Class I MHC incompatible native recipients rejected the allografts after 16.5 days (median) and presensitized recipients after 9 days (median). Concerning class II MHC incompatibility rejection appeared in naive recipients after 16 days (median) and in presensitized recipients after 7 days (median). Hyperacute rejection, however, was not observed. DISCUSSION: These results show an accelerated pattern of rejection in all instances caused by donor MHC antigens. Hyperacute rejection, however, was not observed. The effect is slightly pronounced in class II MHC incompatibility. Acceleration of rejection may be caused by stimulation of the humoral as well as the cellular pathway of the immune response. The underlying mechanisms concerning the different MHC incompatibilities, however, remain speculative.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/imunologia , Transplante de Pâncreas/imunologia , Animais , Anticorpos/sangue , Formação de Anticorpos , Glicemia/metabolismo , Modelos Animais de Doenças , Teste de Histocompatibilidade , Isoanticorpos/sangue , Transplante de Pâncreas/patologia , Transplante de Pâncreas/fisiologia , Ratos , Ratos Endogâmicos , Transplante de Pele/imunologia
9.
Diabetologia ; 45(6): 851-5, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12107729

RESUMO

AIMS/HYPOTHESIS: Thyroid hormones reduce glucose tolerance in animals and humans. This effect is accompanied by a reduction in the beta-cell volume of the pancreas. METHODS: We studied the underlying mechanism using terminal UTP nick end labelling (TUNEL) and caspase-3 to analyse apoptosis and BrdU labelling of beta-cell proliferation. RESULTS: The reason for the reduction of the beta-cell volume of the pancreas after thyroxine treatment is apparently an increased rate of beta-cell apoptosis by an increase of TUNEL and caspase-3 positive rat beta cells. In parallel, thyroxine treatment increased the rate of apoptosis in rat pancreatic ductal cells which are considered to contribute to the pool of stem cells from which insulin-producing beta cells originate. The effects of thyroid hormone treatment are reversible through an increase of the beta-cell replication rate when thyroxine is withdrawn as documented by an increase of the BrdU labelling index. CONCLUSION/INTERPRETATION: An increased rate of beta-cell death due to apoptosis causes a decrease of insulin content and glucose-induced insulin secretion from the pancreas in hyperthyroidism. The resulting reduction of beta cells in the pancreas can provide an explanation for the decrease of glucose tolerance in hyperthyroidism.


Assuntos
Apoptose/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Tiroxina/farmacologia , Animais , Caspase 3 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ratos
10.
Diabetologia ; 44(9): 1189-96, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11596676

RESUMO

AIMS/HYPOTHESIS: We describe a new Type I (insulin-dependent) diabetes mellitus rat model (LEW.1AR1/Ztm-iddm) which arose through a spontaneous mutation in a congenic Lewis rat strain with a defined MHC haplotype (RT1.Aa B/Du Cu). METHODS: The development of diabetes was characterised using biochemical, immunological and morphological methods. RESULTS: Diabetes appeared in the rats with an incidence of 20 % without major sex preference at 58+/-2 days. The disease was characterised by hyperglycaemia, glycosuria, ketonuria and polyuria. In peripheral blood, the proportion of T lymphocytes was in the normal range expressing the RT6.1 differentiation antigen. Islets were heavily infiltrated with B and T lymphocytes, macrophages and NK cells with beta cells rapidly destroyed through apoptosis in areas of insulitis. CONCLUSION/INTERPRETATION: This Type I diabetic rat develops a spontaneous insulin-dependent autoimmune diabetes through beta cell apoptosis. It could prove to be a valuable new animal model for clarifying the mechanisms involved in the development of autoimmune diabetes.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Animais , Apoptose , Linfócitos B/patologia , Glicemia/análise , Diabetes Mellitus Tipo 1/metabolismo , Glicosúria , Hiperglicemia , Insulina/sangue , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Corpos Cetônicos/urina , Células Matadoras Naturais/patologia , Macrófagos/patologia , Masculino , Microscopia Eletrônica , Poliúria , Ratos , Ratos Endogâmicos Lew , Ratos Mutantes , Linfócitos T/patologia
11.
Pancreas ; 23(2): 186-96, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11484921

RESUMO

In the well-established, high-dose streptozotocin diabetic rat model, it is unknown whether normoglycemia after pancreas or islet transplantation may induce the expression of the glucose recognition structures and stimulate the replication of the few surviving pancreatic beta cells. Therefore, the endocrine pancreatic tissue was examined immunocytochemically in streptozotocin-treated major histocompatibility complex congenic Lewis rats at 10 and 100 days after transplantation of whole pancreata or isolated islets implanted under the kidney capsule. In the diabetic state the pancreatic beta cells displayed a weak immunostaining for insulin and glucokinase together with a lack of GLUT2 glucose transporter immunoreactivity in the plasma membrane. Ten days after transplantation, the surviving beta cells had regained their normal immunostaining for insulin and for the glucose recognition structures glucokinase and the A single high dose of streptozotocin causes severe experimental insulin-dependent diabetes mellitus in adult rats due to a selective destruction of the pancreatic beta cells in the islets of Langerhans. At doses between 50 and 60 mg/kg of body weight, only very few beta cells survive in the pancreas (1-3). The diabetic state is irreversible and insulin-dependent, thus representing an experimental animal model for type I diabetes (2). Because of the prevailing hyperglycemia, even the few residual beta cells in the pancreas do not function properly and therefore cannot contribute even to a basal supply of insulin to the organism (4). Pancreatic beta cells can function properly in a diabetic organism apparently only after restitution of normoglycemia (5). GLUT2 glucose transporter. One hundred days after transplantation, both of whole pancreas or isolated islets, the number of surviving beta cells in islets of the pancreata of the recipient animals had increased by two- to threefold. The regenerated beta cells were surrounded by a rim of other endocrine cells. The increase in the number of beta cells was not accompanied by signs of neogenesis from ductal structures in the pancreata. The authors' observations support the concept that strict long-term maintenance of normoglycemia through adequate supply of insulin from endocrine grafts is the ideal prerequisite for beta-cell recovery and restitution of the glucose recognition structures, as well as replication of beta cells in pancreata with end-stage diabetic beta-cell destruction after high-dose streptozotocin treatment.


Assuntos
Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/cirurgia , Transplante das Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/patologia , Transplante de Pâncreas/patologia , Animais , Glicemia/metabolismo , Divisão Celular , Diabetes Mellitus Experimental/sangue , Glucoquinase/metabolismo , Transportador de Glucose Tipo 2 , Imuno-Histoquímica , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Proteínas de Transporte de Monossacarídeos/metabolismo , Ratos , Ratos Endogâmicos Lew , Fatores de Tempo
12.
Xenotransplantation ; 8(1): 41-7, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11208190

RESUMO

Liver xenografts transplanted from guinea pig to rat suffer from inadequate organ reperfusion and initial dysfunction, despite sufficient complement depletion using cobra venom factor (CVF). Reperfusion injury is prevented when complement depleted donors are treated with the prostacyclin analog epoprostenol. Histological analysis suggests that epoprostenol preconditioning prevents post-reperfusion spasms of the intrahepatic branches of the portal vein and strongly reduces appearance of hepatocyte apoptosis shortly after transplantation. Cobra-venom-treated rats show breakdown of glucose metabolism and die in acute hypoglycaemia, whereas the additional application of epoprostenol restores gluconeogenesis. Consequently, recipient survival after epoprostenol and CVF treatment is significantly improved compared with animals receiving CVF only (5.1 +/- 2.6 h vs. 17.9 +/- 5.1 h). These data demonstrate that initial dysfunction of discordant liver grafts in the guinea-pig-to-rat species combination, can be overcome by the application of epoprostenol combined with CVF. Using this pharmacologic regimen, the discordant guinea-pig-to-rat model appears useful to study further questions concerning functional and immunological compatibility of a discordant liver xenograft.


Assuntos
Proteínas Inativadoras do Complemento/administração & dosagem , Venenos Elapídicos/administração & dosagem , Epoprostenol/administração & dosagem , Rejeição de Enxerto/prevenção & controle , Transplante de Fígado , Traumatismo por Reperfusão/prevenção & controle , Animais , Cobaias , Fígado/irrigação sanguínea , Fígado/patologia , Ratos , Transplante Heterólogo
13.
Biochem Pharmacol ; 62(12): 1685-94, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11755122

RESUMO

The desensitization of pancreatic B-cells against stimulation by insulin secretagogues that inhibit ATP-dependent K(+) channels (K(ATP) channels) was investigated by measuring insulin secretion of perifused pancreatic islets. Additionally, the islet insulin content and the number of secretory granules per B-cell were determined. Prior to the measurement of secretion, islets were cultured for 18 h in the presence or absence of the test agents in a cell-culture medium containing 5 mM glucose. The effects of three imidazolines, phentolamine, alinidine, and idazoxan (100 microM each) were compared with those of the well-characterized sulfonylurea, tolbutamide (500 microM), and those of the ion channel-blocking alkaloid, quinine (100 microM). Insulin secretion was strongly reduced upon re-exposure to phentolamine, alinidine, tolbutamide, and quinine, whereas idazoxan, which stimulated secretion only weakly, had no significant effect. The imidazoline secretagogues phentolamine and alinidine induced a cross-desensitization against the stimulatory effect of tolbutamide and quinine. A long-term depolarization with 40 mM KCl was also able to induce a significant reduction of the secretory response to all of the above secretagogues. The insulin content of cultured islets was moderately, but significantly reduced by alinidine, whereas the reduction by phentolamine, tolbutamide, and quinine was not significant. In contrast to these observations, the ultrastructural examination revealed that tolbutamide-treated B-cells had a high degree of degranulation, whereas the other test agents and 40 mM KCl produced only a partial degranulation, except for phentolamine, which produced no significant degranulation at all. These results suggest that the desensitization of insulin secretion is a common property of all agents that stimulate insulin secretion by depolarisation of the plasma membrane. Depending on the specific secretagogue, additional mechanisms, proximal and distal to Ca(2+) influx, appear to contribute to the desensitization (see Rustenbeck et al., pages 1695-1703, this issue).


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Fentolamina/farmacologia , Quinina/farmacologia , Tolbutamida/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Meios de Cultura , Interações Medicamentosas , Hipoglicemiantes/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Potássio/farmacologia
15.
Free Radic Biol Med ; 26(9-10): 1300-4, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10381203

RESUMO

The effect of superoxide dismutase, catalase, metal-chelating agents and hydroxyl radical scavengers on the toxicity of alloxan to isolated ob/ob mouse pancreatic islets in vitro has been compared with the reported ability of such substances to protect against alloxan diabetes in vivo. Superoxide dismutase and catalase protected beta-cells of isolated pancreatic islets against alloxan cytotoxicity, as did the hydroxyl radical scavengers dimethyl sulfoxide (DMSO) and butanol. However, 1,3-dimethylurea and thiourea, that are recognised as effective hydroxyl radical scavengers and that protect animals against the diabetogenic effects of alloxan, were without effect. Similarly, desferrioxamine, that inhibits hydroxyl radical formation from alloxan in chemically defined systems, did not protect against alloxan toxicity. Diethylenetriamine pentaacetic acid, which does not inhibit hydroxyl radical formation from alloxan, also gave no significant protection. The results indicate a role for superoxide radical and hydrogen peroxide in the mechanism of toxicity of alloxan but do not support the involvement of the hydroxyl radical in this process. Alternative explanations must be sought for the ability of hydroxyl radical scavengers and metal-chelating agents to protect against alloxan toxicity in vivo.


Assuntos
Aloxano/toxicidade , Antioxidantes/farmacologia , Quelantes/farmacologia , Sequestradores de Radicais Livres/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , 1-Butanol/farmacologia , Animais , Catalase/farmacologia , Desferroxamina/farmacologia , Diabetes Mellitus Experimental/prevenção & controle , Dimetil Sulfóxido/farmacologia , Radical Hidroxila/metabolismo , Técnicas In Vitro , Ilhotas Pancreáticas/lesões , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Obesos , Superóxido Dismutase/farmacologia
16.
Virchows Arch ; 434(1): 75-82, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10071239

RESUMO

Functional heterogeneity among pancreatic beta cells is a characteristic feature of the islets of Langerhans. Under physiological conditions, beta cells in the pancreas of fed rats exhibited heterogeneous immunohistochemical staining for insulin and glucokinase. Intracellular beta cell glucokinase staining was either faint or dense. In the pericapillary space beta cell glucokinase immunoreactivity had a polar orientation, with the highest density in cytoplasmic regions close to the blood vessels. Starvation resulted in a loss of heterogeneity with homogeneous insulin staining in all beta cells of the islets, and this was accompanied by a loss of heterogeneous glucokinase staining. The intracellular polarity of glucokinase staining in contact to blood vessels also disappeared after starvation. Refeeding resulted in the reappearance of intercellular heterogeneity. In dependence on the functional demand, the endocrine pancreas recruited insulin from beta cells according to a well-defined hierarchy, with an initial preferential mobilization of medullary beta cells. In the course of this process intracellular polarity of glucokinase staining reappeared in areas of the beta cell with functional contact to the GLUT2 glucose transporter in the plasma membrane. This can be regarded as the morphological correlate of an activation of the glucose signal recognition apparatus. Interestingly, the study also provides evidence that the changes in glucokinase distribution apparently preceded those in insulin distribution, which is in keeping with the central role of glucokinase as the glucose sensor of the pancreatic beta cell.


Assuntos
Glucoquinase/análise , Insulina/análise , Ilhotas Pancreáticas/química , Animais , Jejum , Glucoquinase/imunologia , Transportador de Glucose Tipo 2 , Insulina/imunologia , Masculino , Proteínas de Transporte de Monossacarídeos/análise , Ratos , Ratos Wistar
18.
Transfusion ; 37(2): 226-30, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9051101

RESUMO

BACKGROUND: Donor-specific blood transfusion prior to transplantation has been demonstrated to prolong allograft survival. This study compared the effect of donor-specific blood transfusion after kidney, heart, pancreas, and skin allotransplantation in congenic rats of reciprocal strain combinations across a major histocompatibility complex barrier. STUDY DESIGN AND METHODS: Whole donor-specific and third-party blood was administered to the prospective graft recipients 14 and 7 days before the transplantation of kidney, heart, pancreas, and skin. Rejection was defined by cessation of organ-specific function (i.e., uremia, cessation of heartbeat, and recurrence of diabetes). The survival of skin grafts was monitored visually. All allografts were histologically assessed immediately after rejection or at the end of a 100-day observation period. RESULTS: Donor-specific blood transfusion leads to permanent acceptance of all renal allografts. LEW.1U heart allografts were accepted by LEW.1A recipients, but they were rejected in the reciprocal situation. Survival of pancreas and skin allografts was not significantly prolonged. Third-party blood had no effect on allograft survival. CONCLUSION: The beneficial effect of donor-specific blood transfusion depends on both the kind of transplanted organ and the genetic incompatibility involved. The precise mechanisms responsible for the remarkable organ-specific differences remain unknown.


Assuntos
Doadores de Sangue , Transfusão de Sangue , Complexo Principal de Histocompatibilidade , Transplante de Órgãos/fisiologia , Animais , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Transplante de Rim/imunologia , Transplante de Pâncreas/imunologia , Ratos , Transplante de Pele/imunologia , Condicionamento Pré-Transplante
19.
J Endocrinol ; 155(2): 283-93, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9415063

RESUMO

The in vitro toxicity of the diabetogenic agent alloxan as documented by the induction of beta cell necrosis was studied in isolated ob/ob mouse pancreatic islets. The effect of alloxan has been compared with that of a number of N-alkyl alloxan derivatives and with that of the structurally related compound, ninhydrin. Alloxan and its derivatives were selectively toxic to pancreatic beta cells, with other endocrine cells and exocrine parenchymal cells being well preserved, even at high concentration. In contrast, ninhydrin was selectively toxic to pancreatic beta cells only at comparatively low concentration, destroying all islet cell types at high concentrations. The ultrastructural changes induced by all the test compounds in pancreatic beta cells in vitro were very similar to those observed during the development of alloxan diabetes in vivo. The relative toxicity of the various compounds to pancreatic beta cells in vitro was not, however, related to their ability to cause diabetes in vivo. Indeed, the non-diabetogenic substances ninhydrin, N-butylalloxan and N-isobutylalloxan were very much more toxic to isolated islets than the diabetogenic compounds alloxan and N-methylalloxan. These results suggest that the differences in diabetogenicity among alloxan derivatives are not due to intrinsic differences in the susceptibility of the pancreatic beta cells to their toxicity, but may reflect differences in distribution or metabolism. High concentrations of glucose protected islets against the harmful effects of alloxan and its derivatives, but not those of ninhydrin. Low levels of glucose, and non-carbohydrate nutrients, afforded little protection, indicating that the effect of glucose is not due to the production of reducing equivalents within the cell, 3-O-Methylglucose, which protects against alloan diabetes in vivo, did not protect against alloxan toxicity in vitro. Since 3-O-methylglucose is known to prevent uptake of alloxan by pancreatic beta cells, it appears that uptake of alloxan by the cell is not a prerequisite for the induction of beta cell necrosis.


Assuntos
Aloxano/toxicidade , Indicadores e Reagentes/toxicidade , Ilhotas Pancreáticas/efeitos dos fármacos , Ninidrina/toxicidade , Aloxano/análogos & derivados , Animais , Morte Celular , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/ultraestrutura , Camundongos , Microscopia Eletrônica , Necrose
20.
Virchows Arch ; 428(3): 177-85, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8688972

RESUMO

The diabetic Chinese hamster is a well-established animal model for NIDDM with a defective glucose-induced insulin secretory response. In the pancreas of nondiabetic hamsters, the GLUT2 glucose transporter was localized in the plasma membrane of insulin-positive beta cells. At variance with the rat, immunoreactivity was also detected in the cytoplasm. Other islet cell types were not GLUT2 positive. GLUT2 immunoreactivity was already significantly reduced in beta cells from mildly diabetic animals in spite of a normal insulin immunoreactivity. In severely diabetic animals the majority of the beta cells had lost GLUT2 immunostaining. This observation was confirmed in a Western blot analysis of the GLUT2 protein in isolated pancreatic islets. Only beta cells that were densely immunostained for insulin were still GLUT2 positive. However, around 40% of the beta cells devoid of GLUT2 immunoreactivity were still insulin immunoreactive. Thus, the loss of GLUT2 immunoreactivity, which is an important component of the glucose recognition apparatus of the pancreatic beta cell, is an early indicator of beta cell dysfunction before the development of degenerative lesions or the loss of insulin immunoreactivity. GLUT2 loss may be important in the deterioration of glucose-induced insulin secretion in the diabetic Chinese hamster.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Ilhotas Pancreáticas/química , Proteínas de Transporte de Monossacarídeos/análise , Animais , Western Blotting , Membrana Celular/química , Cricetinae , Cricetulus , Citoplasma/química , Modelos Animais de Doenças , Feminino , Transportador de Glucose Tipo 2 , Imuno-Histoquímica , Ilhotas Pancreáticas/ultraestrutura , Masculino , Microscopia Eletrônica , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...