Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Endocrinol Invest ; 40(11): 1201-1207, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28484994

ABSTRACT

AIMS: The dynamical structure of glucose fluctuation has largely been disregarded in the contemporary management of diabetes. METHODS: In a retrospective study of patients with diabetes, we evaluated the relationship between glucose dynamics, antihyperglycemic therapy, glucose variability, and glucose exposure, while taking into account potential determinants of the complexity index. We used multiscale entropy (MSE) analysis of continuous glucose monitoring data from 131 subjects with type 1 (n = 18), type 2 diabetes (n = 102), and 11 nondiabetic control subjects. We compared the MSE complexity index derived from the glucose time series among the treatment groups, after adjusting for sex, age, diabetes duration, body mass index, and carbohydrate intake. RESULTS: In type 2 diabetic patients who were on a diet or insulin regimen with/without oral agents, the MSE index was significantly lower than in nondiabetic subjects but was lowest in the type 1 diabetes group (p < 0.001). The decline in the MSE complexity across the treatment groups correlated with increasing glucose variability and glucose exposure. Statistically, significant correlations existed between higher MSE complexity indices and better glycemic control. In multivariate regression analysis, the antidiabetic therapy was the most powerful predictor of the MSE (ß = -0.940 ± 0.242, R 2 = 0.306, p < 0.001), whereas the potential confounders failed to contribute. CONCLUSIONS: The loss of dynamical complexity in glucose homeostasis correlates more closely with therapy modalities and glucose variability than with clinical measures of glycemia. Thus, targeting the glucoregulatory system by adequate therapeutic interventions may protect against progressive worsening of diabetes control.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Glycemic Index , Hypoglycemic Agents/therapeutic use , Adult , Aged , Body Mass Index , Cross-Sectional Studies , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Female , Humans , Male , Middle Aged , Retrospective Studies
2.
Comput Methods Programs Biomed ; 102(2): 206-18, 2011 May.
Article in English | MEDLINE | ID: mdl-20621384

ABSTRACT

The model-based Karlsburg Diabetes Management System (KADIS®) has been developed as a patient-focused decision-support tool to provide evidence-based advice for physicians in their daily efforts to optimize metabolic control in diabetes care of their patients on an individualized basis. For this purpose, KADIS® was established in terms of a personalized, interactive in silico simulation procedure, implemented into a problem-related diabetes health care network and evaluated under different conditions by conducting open-label mono- and polycentric trials, and a case-control study, and last but not least, by application in routine diabetes outpatient care. The trial outcomes clearly show that the recommendations provided to the physicians by KADIS® lead to significant improvement of metabolic control. This model-based decision-support system provides an excellent tool to effectively guide physicians in personalized decision-making to achieve optimal metabolic control for their patients.


Subject(s)
Decision Support Techniques , Diabetes Mellitus/therapy , Models, Biological , Blood Glucose/metabolism , Case-Control Studies , Clinical Trials as Topic , Computer Simulation , Cybernetics , Diabetes Mellitus/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/therapy , Health Services/statistics & numerical data , Humans
3.
Horm Metab Res ; 41(2): 137-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19214924

ABSTRACT

Given the importance of glucose variability in the development of diabetic complications, the present study used continuous glucose monitoring (CGM) to determine various indices of glucose variability and to investigate their relationships with conventional measures of chronic sustained hyperglycemia. We examined 53 women and 61 men, aged 36-79 years afflicted with type 2 diabetes for 1-24 years. The following indices of glycemic variability were computed from CGM data sets: mean amplitude of glycemic excursions (MAGE), CGM glucose range, interquartile range (IQR), SD-score, and average daily risk range (ADRR). CGM measurements and self-monitored blood glucose (SMBG) records were used to calculate mean CGM sensor glucose and mean SMBG, respectively. In simple correlation analysis, the indices of glucose variability showed weak correlations with HbA1c: MAGE (r=0.27, p <0.01), CGM glucose range (r=0.21, p <0.05), IQR (r=0.31, p <0.01), SD-score (r=0.34, p<0.001), and ADRR (r=0.24, p<0.05). These indices were found to differ at identical HbA1c among several patients, as reflected by diurnal excursions of different frequency and magnitude. With the exception of ADRR, stronger correlations were found between mean SMBG and the other variability indices (r=0.51-0.63, p<0.01 for all). CGM provides various indices of glycemic variability not captured by conventional measures of glycemic control. Detection of the location and the magnitude of glucose fluctuations by CGM should aid in optimal treatment of glycemic disorders in type 2 diabetes.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 2/therapy , Glycemic Index , Monitoring, Physiologic/methods , Adult , Aged , Blood Glucose Self-Monitoring , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Female , Humans , Male , Middle Aged
4.
Horm Metab Res ; 41(3): 207-12, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18975252

ABSTRACT

Cytokine- and FasL-induced pathways contribute to beta-cell death in type 1 diabetes. It remains unclear, however, whether pro-apoptotic cyto-kines or FasL have more apoptotic impact. Cytokine- and FasL-induced apoptosis were simulated using IL-1beta/IFN-gamma, Super-FasLigand and the beta-cell line NIT-1. The role of caspases was addressed using the general caspase inhibitor ZVAD. Exposure to IL-1beta/IFN-gamma induced NIT-1 cell death. FasL augmented cytokine-induced cell death accompanied by increased caspase-3 activation, DNA fragmentation, and chromatin condensation. However, FasL mediated comparable effects on the mitochondrial transmembrane potential (Deltapsi (m)) and nitrite in cytokine- and untreated cells. The cytokine-induced sequence of apoptotic events was (1) Fas, nitrite, (2) Deltapsi (m), (3) DNA fragmentation, cell death, and (4) chromatin condensation. In the presence of FasL, cell death and chromatin condensation appeared earlier implicating a compression of the apoptotic time course. General caspase inhibition using ZVAD prevented cell death, Deltapsi (m), and DNA fragmentation; however, Fas expression and nitrite were increased. In conclusion, cytokines account for the major part of cell death induced by the simultaneously action of FasL + IL-1beta/IFN-gamma. Caspases are of central importance for beta-cell death.


Subject(s)
Apoptosis/drug effects , Fas Ligand Protein/pharmacology , Insulin-Secreting Cells/physiology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Death/drug effects , Cell Line , Flow Cytometry , Glucose/pharmacology , Humans , Insulin/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects
5.
Diabetes Obes Metab ; 10(10): 850-61, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17970756

ABSTRACT

AIM: Incretin enhancers are a new class of antidiabetic drugs with promising therapeutic potential for type 2 diabetes. Therapeutic intervention in prediabetes is an attractive strategy for preventing or delaying diabetes onset. The aim of the present study was to investigate the therapeutic effects of incretin enhancement on incipient impaired glucose tolerance (iIGT) and manifest IGT (mIGT) using the dipeptidyl peptidase IV (DPP-4) inhibitor P32/98- and fatty Zucker rat (ZR, fa/fa) as a model. METHODS: ZRs were classified into groups with iIGT and mIGT (n = 10 per group). P32/98 (21.61 mg/kg body weight) was administered orally to ZR with iIGT and mIGT once daily for 6 and 3 weeks respectively. Assessments included body weight, morning blood glucose and insulin, oral glucose tolerance test (oGTT; 2 g glucose/kg), plasma parameters and blood glucose day-night profile (DNP). In addition, glucose responsiveness of isolated islets and islet morphology were analysed. RESULTS: P32/98 decreased non-fasting morning blood glucose more effectively in ZR with iIGT than in ZR with mIGT. Compared with study entry, P32/98 improved DNP of blood glucose in ZR with mIGT and nearly normalized DNP in ZR with iIGT. An acute bolus of inhibitor reduced glucose load during oGTT in rats chronically treated with placebo or P32/98. In contrast to placebo-treated rats, rats receiving long-term treatment with P32/98 required less insulin during oGTT. This effect was larger in rats with iIGT vs. rats with mIGT. In isolated pancreatic islets of ZR with mIGT, treatment with P32/98 decreased pancreatic insulin content and increased glucose responsiveness, while the beta-cell volume density was unaffected. P32/98 significantly reduced triglycerides and non-esterified fatty acids. Intestinal growth was comparable between inhibitor- and placebo-treated fatty rats. CONCLUSIONS: Enhancement of incretin with the DPP-4 inhibitor P32/98 has therapeutic effects in hyperinsulinaemia, hyperglycaemia and IGT in ZR with iIGT and mIGT. Apparently, administration of P32/98 in ZR with iIGT results in more efficient beta-cell function, which is associated with less need for insulin to cope with deterioration of glucose tolerance. Importantly, P32/98 has a strong effect on dyslipidaemia in mIGT. P32/98 has no side effect on intestinal growth. Daily intake of P32/98 is a promising strategy for treatment of glucose intolerance and has the potential to prevent type 2 diabetes.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucose Intolerance/drug therapy , Isoleucine/analogs & derivatives , Thiazoles/therapeutic use , Animals , Blood Glucose/analysis , Body Weight/drug effects , Fatty Acids, Nonesterified/blood , Glucose/pharmacology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Incretins/metabolism , Insulin/analysis , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/chemistry , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Isoleucine/therapeutic use , Male , Models, Animal , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Rats , Rats, Zucker , Time Factors , Tissue Culture Techniques , Triglycerides/blood
6.
Diabetes Obes Metab ; 7(2): 170-81, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15715890

ABSTRACT

AIM: The aim of this study was to investigate the effect of long-term treatment with the dipeptidyl peptidase inhibitor P32/98 and its combination with rosiglitazone on blood glucose control and islet of Langerhans histology in male Zucker diabetic fatty (ZDF) rats, when treatment begins before or after the development of overt diabetes. METHODS: ZDF rats were treated with P32/98 from the age of 9, 12 or 15 weeks. Rosiglitazone maleate was given to a separate group from the age of 13 weeks. P32/98 was given to all of these rosiglitazone-treated rats from 16 weeks of age. Rosiglitazone maleate was also given from 16 weeks of age to half the rats that were given P32/98 from 9 weeks of age. The compounds were given by oral gavage until the rats were 14 weeks old and then in the diet. The experiment was terminated at the age of 20-21 weeks. Blood glucose, plasma insulin and oral glucose tolerance were measured at intervals; islet histology was assessed terminally. RESULTS: P32/98 improved glucose tolerance after both single and multiple doses when treatment started at 9 weeks of age, also after the third week of treatment when treatment began at 12 or 15 weeks of age. P32/98 reduced daytime blood glucose when treatment began at 12 weeks. Treatment with rosiglitazone increased food intake and body weight, and after 2 weeks, reduced daytime blood glucose, water intake and the area under the glucose tolerance curve. A single dose of P32/98 markedly improved glucose tolerance in rosiglitazone-treated rats. When treatment had begun at 9 weeks of age, P32/98 stimulated insulin secretion in some glucose tolerance tests. Neither P32/98 nor rosiglitazone affected pancreatic insulin content, nor did they have clear effects on islet histology. CONCLUSION: P32/98 elicited a sustained improvement in glucose tolerance in both prediabetic and diabetic ZDF rats. The effects of P32/98 on glucose and insulin were similar to those of rosiglitazone, and in contrast to rosiglitazone, P32/98 did not increase food intake or body weight. However, neither compound was especially effective at improving diabetes in ZDF rats when treatment began at 9, 12 or 15 (P32/98) or 13 (rosiglitazone) weeks of age.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/metabolism , Hypoglycemic Agents/therapeutic use , Pentanoic Acids/therapeutic use , Thiazoles/therapeutic use , Thiazolidinediones/therapeutic use , Animals , Body Weight , Drinking , Drug Therapy, Combination , Drug Tolerance , Eating/physiology , Glucose Tolerance Test , Insulin/blood , Islets of Langerhans/drug effects , Male , Obesity/drug therapy , Obesity/metabolism , Pentanoic Acids/blood , Rats , Rats, Zucker , Rosiglitazone , Thiazoles/blood , Thiazolidines , Time Factors
7.
Endocrinology ; 145(6): 2747-52, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15033908

ABSTRACT

In the pathogenesis of autoimmune type 1 diabetes, the apoptosis receptor Fas appears de novo on the surface of insulin-producing beta-cells. Fas expression is thought to be induced by proinflammatory cytokines, such as IL-1beta, interferon-gamma (IFNgamma), and TNFalpha, released by islet-infiltrating mononuclear cells. To determine whether beta-cells can modulate their sensitivity to apoptosis at the level of Fas, we investigated the effect of Fas ligand (FasL) on surface expression of Fas in NIT-1 insulinoma cells from nonobese diabetic (NOD) mice prone to autoimmune diabetes and islet cells from NOD and nonautoimmune BALB/c mice. In NIT-1 insulinoma cells, Fas expression induced by the cytokine combination IL-1beta and IFNgamma was reduced in the presence of FasL, whereas in islet cells Fas expression was unaffected by FasL. The effect of FasL on NIT-1 cells was evident during and after the induction of Fas expression by IL-1beta and IFNgamma. Thus, FasL down-regulates cytokine-induced Fas expression in NOD mouse-derived NIT-1 cells, but not in NOD or BALB/c mouse islets. The ability of NIT-1 cells to down-regulate Fas receptor in response to ligation is similar to that of a variety of tumor cells, which may use this mechanism to escape destruction by cytotoxic T cells. Islets apparently cannot protect themselves against FasL-induced apoptosis by down-regulating the Fas receptor. Understanding how NIT-1 insulinoma cells down-regulate Fas receptor in response to ligation by FasL has therapeutic implications for protecting normal beta-cells in autoimmune type 1 diabetes.


Subject(s)
Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulinoma/metabolism , Membrane Glycoproteins/metabolism , Pancreatic Neoplasms/metabolism , fas Receptor/metabolism , Animals , Apoptosis , Cells, Cultured , Diabetes Mellitus, Type 1/pathology , Down-Regulation , Fas Ligand Protein , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Time Factors
8.
J Mol Endocrinol ; 30(2): 163-71, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12683940

ABSTRACT

During the process of insulitis in the pathogenesis of type I (insulin-dependent) diabetes mellitus, proinflammatory cytokines induce expression of the death receptor Fas on the surface of pancreatic beta-cells and thereby contribute to the enhanced susceptibility of beta-cells for apoptosis. The aim of this study was to compare cell-surface and intracellular Fas expression associated with cytokine-induced apoptosis in commonly used beta-cell models such as isolated islets and insulinoma lines derived from mouse and rat. The cell line NIT-1 responded to the interleukin (IL)-1beta+interferon (IFN)-gamma stimulus with translocation of Fas to the cell surface. Likewise, islet cells from non-obese diabetic (NOD) mice and BB/OK rats expressed increasing amounts of the Fas receptor on their surfaces after exposure to IL-1beta in combination with IFN-gamma and tumour necrosis factor-alpha. Moreover, islets obtained from BB/OK rats at an age near the onset of diabetes had an increased surface expression of Fas compared with young rats. In contrast, western blot analysis of cell lysates from cytokine-exposed islets and insulinoma cells revealed total Fas expression levels comparable to those of untreated controls. In conclusion, islets from BB/OK rats and NOD mice, in addition to NIT-1 insulinoma cells, responded to cytokine exposure with surface expression of the Fas receptor, whereas in cell lysates the levels of expression of Fas were found to be independent of cytokine exposure. Taken together, the findings indicate that cytokine-treated beta-cells might possess two pools of Fas protein, one of which is inducible by cytokines and accounts for surface Fas expression, whereas the other is constitutively expressed in cytoplasmic compartments. The underlying mechanisms, including possible interactions between these two sources of cellular Fas expression, need to be investigated in future studies.


Subject(s)
Apoptosis/physiology , Insulinoma/metabolism , Interferon-gamma/metabolism , Interleukin-1/metabolism , Islets of Langerhans/metabolism , fas Receptor/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Insulinoma/immunology , Insulinoma/pathology , Interferon-gamma/immunology , Interleukin-1/immunology , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Nitric Oxide/metabolism , Rats , Rats, Inbred BB , fas Receptor/immunology
9.
Biochem Biophys Res Commun ; 290(1): 443-51, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11779190

ABSTRACT

The appearance of Fas receptor at the surface of pancreatic beta-cells affected by progressive insulitis strongly suggests that Fas-mediated beta-cell apoptosis plays an important role in the pathogenesis of type 1 diabetes. In support of this concept, the present study has shown that islet cells from NOD mice and the beta-cell line NIT-1 respond to the proinflammatory cytokines IL-1beta and IFN-gamma with Fas surface expression in a dose- and time-dependent manner. Moreover, the prevention of cytokine-induced surface Fas expression by actinomycin D, cycloheximide, and brefeldin A demonstrated that trafficking of Fas to the beta-cell surface requires RNA and protein synthesis and, in addition is critically dependent on intracellular protein transport. Compared with total cellular Fas protein, the amount of Fas at the cell surface was relatively small and indicated that Fas is preferentially expressed in cytoplasmic compartments of NIT-1 cells. It is concluded that inflammatory insults specifically induce translocation of Fas to the beta-cell surface and that interference with cell surface Fas expression is a new strategy to improve beta-cell survival in inflamed islets.


Subject(s)
Islets of Langerhans/metabolism , fas Receptor/biosynthesis , Animals , Apoptosis , Blotting, Western , Brefeldin A/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Coculture Techniques , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Diploidy , Dose-Response Relationship, Drug , Flow Cytometry , Inflammation , Interferon-gamma/metabolism , Interleukin-1/metabolism , Mice , Mice, Inbred NOD , Nitric Oxide/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Biosynthesis , Protein Synthesis Inhibitors/pharmacology , Protein Transport , RNA/metabolism , RNA, Messenger/metabolism , Time Factors , Tumor Cells, Cultured
10.
J Inherit Metab Dis ; 24(1): 15-27, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11286378

ABSTRACT

NDUFA1 is one of the 36 nuclear genes encoding subunits of the mitochondrial complex I involved in the respiratory chain. The human NDUFA1 has been cloned, completely sequenced and mapped to Xq24. In the present study, we searched for sequence variations in NDUFA1 as causative defects in complex I deficiency using genomic DNA of 152 patients with various clinical phenotypes. The patient sample consisted of 54 patients (46 male and 8 female) with Leber heriditary optic neuropathy (LHON) from 48 unrelated families from Germany and 98 patients (72 male and 26 female) with biochemically proven complex I deficiency including Leigh syndrome. Patient DNA was used to amplify all three exons, including the exon/intron boundaries and the promoter region of NDUFA1 for heteroduplex analysis and direct sequencing. In the 152 patients tested, no mutation was found that could be related to any of the disease phenotypes included. However, three single-nucleotide polymorphisms (SNPs) located in the promoter region (SNP G/C at nt -71 and SNP T/C at nt -189) and in intron 1 (SNP T/G nt 1454) were discovered. Allele frequencies of the SNPs were estimated in a German and Estonian control population and compared to complex I-deficient patients. There was no significant difference between the control population, the LHON patients, or the severely affected patients with complex I deficiency, excluding an association of the polymorphisms with the diseases. Our results suggest that mutations in NDUFA1 do not cause the gender difference observed in clinically severe and complex phenotypes with complex I deficiency.


Subject(s)
Membrane Proteins/genetics , NADH, NADPH Oxidoreductases/genetics , Alleles , DNA Mutational Analysis , Electron Transport , Electron Transport Complex I , Female , Gene Frequency , Genetic Variation , Humans , Leigh Disease/genetics , Male , NADH Dehydrogenase , NADH, NADPH Oxidoreductases/deficiency , Nucleic Acid Heteroduplexes , Optic Atrophies, Hereditary/genetics , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
11.
Diabetologia ; 41(11): 1381-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9833948

ABSTRACT

Autoimmune-mediated destruction of pancreatic islet beta cells leads to insulin-dependent diabetes in non-obese diabetic (NOD) mice. Although both direct cytotoxic T cell- and indirect cytokine-, nitric oxide- or free radical-mediated mechanisms induce beta-cell apoptosis in vitro, beta-cell death in vivo in spontaneous autoimmune diabetes is not well-characterized. Furthermore, whether beta cells die gradually, or rapidly in the late pre-clinical stage, is a question of current interest. To investigate beta-cell death in vivo, we measured the frequency and intra-islet localisation of apoptosis, defined as DNA strand breaks by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) technique, during spontaneous and cyclophosphamide-accelerated diabetes in NOD mice. In spontaneous diabetes, the frequency of apoptosis in islets correlated with the progression of beta-cell destruction with age. Although apoptosis was detected at low frequency within the reduced insulin-positive islet area of pre-diabetic mice at 90 days of age, it was rarely co-localised to beta cells. After acceleration of beta-cell destruction with cyclophosphamide, the frequency of apoptosis reached maximum at 12 days, at which time 3.2 % of apoptotic cells were beta cells. Apoptosis was most frequent in the insulin-negative islet area comprised of mononuclear cell infiltrate and was localized to CD8+ T cells. The rarity of detectable apoptotic beta cells in spontaneous pre-diabetic mice with pronounced insulitis and reduced insulin-positive islet areas most likely reflects the rapid clearance of apoptotic beta cells. Our findings are more consistent with gradual destruction of non-renewable beta-cells in spontaneous diabetes, than with their rapid, accelerated destruction (as after cyclophosphamide) in the late pre-clinical stage.


Subject(s)
Apoptosis , Cyclophosphamide/pharmacology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Insulin/analysis , Islets of Langerhans/pathology , Aging , Animals , Diabetes Mellitus, Type 1/immunology , Disease Progression , Female , In Situ Nick-End Labeling , Islets of Langerhans/drug effects , Islets of Langerhans/growth & development , Mice , Mice, Inbred NOD , Time Factors
12.
Mol Med ; 4(8): 495-501, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9742505

ABSTRACT

BACKGROUND: The non-obese diabetic (NOD) mouse is a model of human type 1 diabetes in which autoreactive T cells mediate destruction of pancreatic islet beta cells. Although known to be triggered by cytotoxic T cells, apoptosis has not been unequivocally localized to beta cells in spontaneously diabetic NOD mice. We created a model of accelerated beta-cell destruction mediated by T cells from spontaneously diabetic NOD mice to facilitate the direct detection of apoptosis in beta cells. MATERIALS AND METHODS: NOD.scid (severe combined immunodeficiency) mice were crossed with bm1 mice transgenically expressing the costimulatory molecule B7-1 (CD80) in their beta cells, to generate B7-1 NOD.scid mice. Apoptosis in islet cells was measured as DNA strand breakage by the TdT-mediated-dUTP-nick end labeling (TUNEL) technique. RESULTS: Adoptive transfer of splenocytes from spontaneously diabetic NOD mice into B7-1 NOD.scid mice caused diabetes in recipients within 12-16 days. Mononuclear cell infiltration and apoptosis were significantly greater in the islets of B7-1 NOD.scid mice than in nontransgenic NOD.scid mice. Dual immunolabeling for TUNEL and either B-7 or insulin, or the T cell markers CD4 and CD8, and colocalization by confocal microscopy clearly demonstrated apoptosis in beta cells as well in a relatively larger number of infiltrating T cells. The clearance time of apoptotic beta cells was estimated to be less than 6 min. CONCLUSIONS: B7-1 transgenic beta cells undergo apoptosis during their accelerated destruction in response to NOD mouse effector T cells. Rapid clearance implies that beta cells undergoing apoptosis would be detected only rarely during more protracted disease in spontaneously diabetic NOD mice.


Subject(s)
Apoptosis/immunology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Adoptive Transfer , Animals , B7-1 Antigen/physiology , Diabetes Mellitus, Type 1/immunology , Insulin/analysis , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Pancreas/immunology , Pancreas/pathology , Spleen/immunology , T-Lymphocytes/physiology
13.
J Immunoassay ; 18(4): 289-307, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9358337

ABSTRACT

Although various methods for the detection of autoantibodies against glutamic acid decarboxylase (GAD65-AAb) are known, no sensitive method for the quantification of GAD65 as autoantigen is available. We describe a sandwich ELISA based on monoclonal GAD65 antibodies (Mc-GAD65-Ab) of different epitope specificities to quantify GAD65 in pancreatic islets and in different organ/cell extracts and during the preparation of GAD from brain extracts. GAD65 was captured via solid phase coated Mc-GAD65-Ab and detected via a second biotin-labelled Mc-GAD65-Ab recognizing a NH2-terminal epitope of the molecule. The detection limit was estimated to be 0.03 ng GAD65/ml using alkaline phosphatase (AP)-conjugated streptavidin. GAD65 contents in islets of neonatal BB/OK rats and Lewis rats amounted to 37.4 and 43.7 pg/islet, respectively. Furthermore, GAD65 was quantified in brain extracts of pig (55.1 ng/mg protein), mouse (39.5 ng/mg), rat (243.8 ng/mg) and pig cerebellum (514.8 ng/mg) and in different organ extracts of Lewis rat.


Subject(s)
Antibodies, Monoclonal , Autoantibodies/analysis , Autoantigens/analysis , Diabetes Mellitus, Type 1/enzymology , Enzyme-Linked Immunosorbent Assay/methods , Glutamate Decarboxylase/analysis , Isoenzymes/analysis , Animals , Brain Chemistry , Cell Line , Diabetes Mellitus, Type 1/immunology , Epitope Mapping , Female , Glutamate Decarboxylase/immunology , Humans , Islets of Langerhans/chemistry , Isoenzymes/immunology , Male , Mice , Mice, Inbred BALB C , Molecular Weight , Rats , Rats, Inbred Lew , Recombinant Proteins/analysis , Sensitivity and Specificity
14.
Pancreas ; 15(2): 139-46, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9260198

ABSTRACT

GAD65 is targeted by different patterns of autoantibodies [glutamic acid decarboxylase (GAD)-AAbs] in insulin-dependent diabetes mellitus (IDDM) and stiff-man syndrome (SMS). To study differentiation of the GAD-AAb pattern by immunohistochemistry, we examined the immunostaining of 15 monoclonal GAD antibodies (mc-GAD-Abs), which recognized different epitope regions of the antigen, on human pancreatic sections that were unfixed or fixed with different fixatives. By a competitive sandwich enzyme-linked immunosorbent assay (ELISA), three binding patterns of mc-GAD-Abs were identified: 5 of 15 mc-GAD-Abs recognized a linear N-terminal epitope (p1), 5 of 15 were reactive with a conformational GAD65 epitope region (p2), and 5 of 15 were cross-reactive with GAD67 (p3). These patterns of mc-GAD-Abs were tested for islet cell binding by indirect immunofluorescence on pancreatic sections treated with either (1) Bouin's solution, (2) Zamboni's solution, or (3) phosphate-buffered formaldehyde for 0.5, 1, 2, and 18 h at 4 degrees C. After fixation for up to 2 h no differentiation of immunoreactivity of patterns was observed using the three fixatives. mc-GAD-Abs recognizing conformational epitope regions (p2) revealed a marked reduced immunoreactivity on pancreatic sections fixed for 18 h with 4% formaldehyde, while mc-GAD-Abs reactive with linear epitopes (p1, p3) were detectable with strong binding. This fixation procedure was used to compare the immunoreactivity of GAD-AAb+ or GAD-AAb- islet cell cytoplasmic antibody-positive (ICA+) sera of IDDM (n = 27) and SMS patients (n = 3). The three SMS sera were reactive with GAD on fixed islets but showed a reduced titer, whereas the majority of IDDM sera (22/27; 81.5%) were not detectable; 70.6% (12/ 17) of GAD-AAb+ IDDM sera were not detectable on fixed islets. Furthermore, all 10 GAD-AAb- IDDM sera tested failed to react with fixed pancreas, which also suggested an alteration of non-GAD-ICA antigens. In conclusion, the fixation of human pancreatic sections with formaldehyde for 18 h allows the differentiation of GAD-AAbs recognizing linear and conformational epitope regions.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Epitopes/chemistry , Epitopes/immunology , Glutamate Decarboxylase/immunology , Immunohistochemistry , Animals , Antibodies, Monoclonal/immunology , Binding, Competitive , Diabetes Mellitus, Type 1/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Glutamate Decarboxylase/chemistry , Humans , Mice , Mice, Inbred BALB C , Pancreas/enzymology , Protein Conformation , Recombinant Proteins
15.
J Exp Med ; 185(6): 1013-21, 1997 Mar 17.
Article in English | MEDLINE | ID: mdl-9091575

ABSTRACT

The class II major histocompatibility complex molecule I-A(g7) is strongly linked to the development of spontaneous insulin-dependent diabetes mellitus (IDDM) in non obese diabetic mice and to the induction of experimental allergic encephalomyelitis in Biozzi AB/H mice. Structurally, it resembles the HLA-DQ molecules associated with human IDDM, in having a non-Asp residue at position 57 in its beta chain. To identify the requirements for peptide binding to I-A(g7) and thereby potentially pathogenic T cell epitopes, we analyzed a known I-A(g7)-restricted T cell epitope, hen egg white lysozyme (HEL) amino acids 9-27. NH2- and COOH-terminal truncations demonstrated that the minimal epitope for activation of the T cell hybridoma 2D12.1 was M12-R21 and the minimum sequence for direct binding to purified I-A(g7) M12-Y20/K13-R21. Alanine (A) scanning revealed two primary anchors for binding at relative positions (p) 6 (L) and 9 (Y) in the HEL epitope. The critical role of both anchors was demonstrated by incorporating L and Y in poly(A) backbones at the same relative positions as in the HEL epitope. Well-tolerated, weakly tolerated, and nontolerated residues were identified by analyzing the binding of peptides containing multiple substitutions at individual positions. Optimally, p6 was a large, hydrophobic residue (L, I, V, M), whereas p9 was aromatic and hydrophobic (Y or F) or positively charged (K, R). Specific residues were not tolerated at these and some other positions. A motif for binding to I-A(g7) deduced from analysis of the model HEL epitope was present in 27/30 (90%) of peptides reported to be I-A(g7)-restricted T cell epitopes or eluted from I-A(g7). Scanning a set of overlapping peptides encompassing human proinsulin revealed the motif in 6/6 good binders (sensitivity = 100%) and 4/13 weak or non-binders (specificity = 70%). This motif should facilitate identification of autoantigenic epitopes relevant to the pathogenesis and immunotherapy of IDDM.


Subject(s)
Genes, MHC Class II , Histocompatibility Antigens Class II/chemistry , Proinsulin/chemistry , Amino Acid Sequence , Animals , Binding Sites , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Epitopes/analysis , Histocompatibility Antigens Class II/genetics , Humans , Hybridomas , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Inbred Strains , Molecular Sequence Data , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology
16.
Acta Diabetol ; 33(3): 225-31, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8904930

ABSTRACT

To study the immune response to glutamic acid decarboxylase (GAD) in insulin-dependent diabetes mellitus, monoclonal GAD antibodies after fusion of splenocytes from a nondiabetes-susceptible BALB/c mouse immunized with human recombinant GAD65 were generated. Of the 44 monoclonals, 35 are specific for the GAD65 isoform, whereas 9 also react with GAD67. Some 37 monoclonals, including all GAD65/67 reactive antibodies, react with GAD by Western blot analysis. The remaining 7 GAD65 monoclonals bind GAD only in an immunoprecipitation assay, which implies that they target epitopes dependent on the conformation of the GAD molecule. The 125I-GAD binding of the GAD65 monoclonals reactive on Western blotting was significantly diminished by all 3 sera from Stiff-man syndrome patients but only by 3/30 (10%) sera from type 1 diabetic patients. In contrast, the 7 monoclonal antibodies reactive with a conformation-dependent GAD epitope were competitive with 83% of GAD-autoantibody-positive sera from these diabetic patients. Using chimeric GAD65/67 proteins, the epitope region targeted by these monoclonals was mapped to the middle of GAD65 (amino acids 221-442). This central conformation-dependent GAD region was also targeted by sera from patients with type 1 diabetes. In conclusion, our data show that even after common immunization of a nondiabetes-susceptible mouse strain, monoclonal were obtained which preferentially react with the GAD65 linear amino-terminus (amino acids 4-17) and a conformation-dependent region located in the middle of GAD targeted by autoantibodies, indicating that this GAD region is not restricted to the autoimmune response associated with the Stiff-man syndrome and the beta-cell destruction in type 1 diabetes mellitus.


Subject(s)
Antibodies, Monoclonal , Autoantibodies/blood , Diabetes Mellitus, Type 1/immunology , Epitopes/analysis , Glutamate Decarboxylase/immunology , Stiff-Person Syndrome/immunology , Animals , Brain/enzymology , Diabetes Mellitus, Type 1/blood , Disease Susceptibility , Epitopes/immunology , Female , Fluorescent Antibody Technique, Indirect , Humans , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Mice , Mice, Inbred BALB C/immunology , Protein Conformation , Rats , Recombinant Proteins/immunology , Stiff-Person Syndrome/blood
17.
J Autoimmun ; 9(3): 309-13, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8816965

ABSTRACT

To study the ability of interleukin 1-beta (IL-1) to induce apoptosis in the endocrine pancreas, rat islets of Langerhans obtained from 14-day-old BB.1A rats were exposed to 25 U/ml IL-1 for 40 h. In order to prove the role of nitric oxide (NO) in this process islets were exposed either to 1 mmol/l N-nitro-L-arginine methylester (NAME) or to 25 mmol/l nicotinamide (NA) or to a combination of NA or NAME with IL-1. In dispersed cells oligonucleosomes, resulting from cleavage of nuclear DNA due to apoptosis, were identified by enzymatic labelling the free 3'-OH-DNA ends with fluorescein-dUTP and quantified by means of flow cytometry. After exposure to IL-1, the islets were characterized by elevated basal (in response to 2 mmol/l glucose) insulin release while glucose-stimulated (20 mmol/l glucose) insulin secretion was nearly completely abolished. In contrast, glucose-stimulated insulin secretion was well preserved in NAME-exposed islets, but was markedly inhibited after NA treatment. Accordingly, only the IL-1-induced inhibition of glucose-stimulated insulin secretion was significantly restored in the presence of NAME but was reinforced by NA. IL-1 exposure resulted in a significant increase in the percentage of apoptotic cells (untreated controls 3.8 +/- 0.5% IL-1 18.8 +/- 1.8%, P < 0.01). This effect was significantly reduced in the presence of NA and NAME. Nitrite production which was assayed as an equivalent of NO generation of islets was highest under the influence of IL-1 (16.48 +/- 1.40 versus 2.89 +/- 0.37 pmol/islet for control islets) which was correlated with the percentage of apoptotic cells. IL-1-stimulated nitrite production was reduced to 9.25 +/- 0.48 and 3.41 +/- 0.36 pmol/islet in the presence of NA or NAME, respectively. The results demonstrate the potency of IL-1 to induce apoptosis in rat islets. Since inhibition of NO production was always paralleled by a reduced ability of IL-1 to induce programmed cell death, this radical appears to be involved in this process. Remarkably, the near-complete prevention of NO generation as demonstrated under the influence of NAME was able to prevent the IL-1-induced deterioration of glucose-stimulated insulin secretion in parallel to the prevention of apoptosis-related appearance of DNA double-strand breaks. It is concluded that the elimination of damaged beta cells due to IL-1 exposure is partly achieved by induction of apoptosis.


Subject(s)
Apoptosis/drug effects , Interleukin-1/pharmacology , Islets of Langerhans/cytology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , DNA/drug effects , DNA Damage , Enzyme Inhibitors/pharmacology , Insulin/metabolism , NG-Nitroarginine Methyl Ester , Niacinamide/pharmacology , Nitric Oxide/biosynthesis , Rats , Rats, Inbred BB
18.
Acta Histochem ; 98(2): 229-41, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8739307

ABSTRACT

In order to compare the reactivity of glutamic acid decarboxylase (GAD) antibodies recognizing linear and conformational epitopes as islet cell cytoplasmic antibodies (ICA), monoclonal antibodies were generated. An ELISA displacement test using two biotinylated monoclonals recognizing a linear (M61/7E11) or a conformational GAD65 epitope (M65/6B12) was performed to identify epitope regions recognized by monoclonal GAD antibodies. The GAD binding by monoclonal GAD antibodies was tested by immunofluorescence on fixed and unfixed pancreatic sections of human, rat, and mouse, and by Dot-blot experiments. 16/23 (69.6%) of the monoclonals were specifically reactive with GAD65 and 7/23 (30.4%) were reactive with both GAD isoforms. 8/16 (50%) of monoclonal GAD65 antibodies recognized a linear GAD epitope located at the N-terminus (pattern 1). 5/16 (31.3%) displaced M65/6B12, indicating the recognition of a conformational GAD epitope (pattern 2). Monoclonals belonging to patterns 1 and 2 showed strong ICA binding. 3/16 (18.8%) of monoclonals specific for GAD65 with weak or no immunostaining of pancreatic islets (pattern 3) did not inhibit the binding of both biotinylated antibodies in the displacement test, indicating other epitope specificities. In conclusion, GAD antibodies recognizing both conformational and linear epitopes of the GAD65 molecule are involved in ICA binding with strong reactivity. Furthermore, results obtained with monoclonals of pattern 3 suggest the occurrence of GAD65 epitopes partly inaccessible on cryosections, which may result in an ICA-negative test of GAD65 autoantibody positive sera.


Subject(s)
Antibodies, Monoclonal/immunology , Autoantibodies/immunology , Epitopes/immunology , Glutamate Decarboxylase/chemistry , Islets of Langerhans/immunology , Animals , Blotting, Western , Cerebellum/enzymology , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Fluorescent Antibody Technique , Glutamate Decarboxylase/immunology , Glutamate Decarboxylase/metabolism , Humans , Islets of Langerhans/enzymology , Mice , Mice, Inbred BALB C , Pancreas/cytology , Pancreas/enzymology , Protein Conformation , Rats , Rats, Inbred Strains
19.
Horm Metab Res ; 28(1): 11-5, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8820987

ABSTRACT

After fusion of splenocytes from a Balb/c mouse which received three injections of human recombinant GAD65 44 hybridomas producing monoclonal GAD antibodies (mc-GAD Ab) could be established. 35 out of the 44 mc-GAD Ab specifically recognized the GAD65 isoform, whereas 9 showed a cross-reactivity with GAD67. All antibodies belong to the IgG class. The mc-GAD Ab were reactive with recombinant as well as natural GAD tested in enzyme-linked immunosorbent assay. Twenty-one antibodies stained islet cells very well in cryosections of human, monkey, rat, and pig pancreas. However, the mc-GAD Ab failed to bind on the surface of viable rat islet cells, although they reveal a striking binding on permeabilized cells examined by cytofluorometry. Furthermore, the mc-GAD Ab were analyzed for complement-dependent antibody-mediated cytoxicity (C'AMC) to rat islet cells. Whereas our monoclonal Beta-cell specific surface antibody K14D10 caused a high C'AMC measured as a 51Cr-release of 56.5 +/- 4.6%, n = 10, only 4/44 mc-GAD Ab provoked a moderate increase of 51Cr-release ranging from 7.1-10.5% (cut-off 7.0%). From these findings it is suggested that GAD is not detectable on the islet-cell surface by binding and cytotoxicity test.


Subject(s)
Glutamate Decarboxylase/metabolism , Islets of Langerhans/enzymology , Membrane Glycoproteins/metabolism , Animals , Antibodies, Monoclonal , Antibody Specificity , Chromium Radioisotopes , Complement System Proteins/immunology , Female , Fluorescent Antibody Technique, Indirect , Glutamate Decarboxylase/immunology , Humans , Hybridomas/immunology , Immunoglobulin G/immunology , Islets of Langerhans/cytology , Mice , Mice, Inbred BALB C , Rats
20.
Acta Diabetol ; 32(4): 217-24, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8750759

ABSTRACT

The influence of beta cell activity on cytokine-induced functional and structural impairments as well as the ability of those damaged cells to recover were investigated. Rat islets cultured for 4 days in the presence of 5, 10, and 30 mmol/l glucose were exposed to interferon-gamma (IFN, 500 U/ml) and tumor necrosis factor-alpha (TNF, 250 U/ml) for the last 24 h. After cytokine removal islets were allowed to recover spontaneously in culture medium containing 10 mmol/l glucose for a further 7 days. Cytokines significantly inhibited insulin release into culture medium, insulin storage, glucose-stimulated insulin secretion, protein, and DNA synthesis. In the presence of cytokines there was a six- to eightfold increase in nitrite production by the islets. The functional impairments were more pronounced in metabolically stimulated beta cells. In addition, cytokines caused membrane alterations as indicated by increased spontaneous chromium-51 release. The cytokines specifically induced the synthesis of two proteins (72 and 88 kDa, respectively). By immunoblotting, the 72-kDa protein was identified as heat shock protein. After a 1-week recovery period, insulin storage and stimulated insulin secretion of cytokine-treated islets were still significantly diminished. However, protein and DNA synthesis of cytokine-exposed islets returned to pre-exposure levels. In conclusion, high beta cell activity increases islet susceptibility to TNF+IFN. Cytokine-induced, long-lasting, inhibitory effects are primarily directed to beta-cell-specific functions, while general vital cell functions clearly recover after cytokine removal. The induction of certain proteins and the increased protein synthesis and replication rate after cytokine removal might reflect activated repair processes.


Subject(s)
Cytokines/toxicity , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Interferon-gamma/toxicity , Islets of Langerhans/drug effects , Kinetics , Rats , Rats, Inbred BB , Recombinant Proteins/toxicity , Time Factors , Tumor Necrosis Factor-alpha/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...