Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters








Publication year range
1.
Am J Transplant ; 13(9): 2308-21, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23855618

ABSTRACT

To ensure safety tolerance induction protocols are accompanied by conventional immunosuppressive drugs (IS). But IS such as calcineurin inhibitors (CNI), for example, cyclosporin A (CsA), can interfere with tolerance induction. We investigated the effect of an additional transient CsA treatment on anti-CD4mAb-induced tolerance induction upon rat kidney transplantation. Additional CsA treatment induced deteriorated graft function, resulting in chronic rejection characterized by glomerulosclerosis, interstitial fibrosis, tubular atrophy and vascular changes. Microarray analysis revealed enhanced intragraft expression of the B cell attracting chemokine CXCL13 early during CsA treatment. Increase in CXCL13 expression is accompanied by enhanced B cell infiltration with local and systemic IgG production and C3d deposition as early as 5 days upon CsA withdrawal. Adding different CNIs to cultures of primary mesangial cells isolated from glomeruli resulted in a concentration-dependent increase in CXCL13 transcription. CsA in synergy with TNF-α can enhance the B cell attracting and activating potential of mesangial cells. Transient B cell depletion or transfer of splenocytes from tolerant recipients 3 weeks after transplantation could rescue tolerance induction and did inhibit intragraft B cell accumulation, alloantibody production and ameliorate chronic rejection.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , Calcineurin Inhibitors , Immune Tolerance/immunology , Immunosuppressive Agents/pharmacology , Kidney Transplantation , Animals , B-Lymphocytes/immunology , Calcineurin/pharmacology , Chemokine CXCL13/biosynthesis , Cyclosporine/pharmacology , Graft Rejection/drug therapy , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immune Tolerance/drug effects , Kidney/metabolism , Lymphocyte Activation , Male , Rats , Rats, Inbred Lew
2.
Am J Transplant ; 12(9): 2384-94, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22702307

ABSTRACT

Recent data suggest that donor-specific memory T cells (T(mem)) are an independent risk factor for rejection and poor graft function in patients and a major challenge for immunosuppression minimizing strategies. Many tolerance induction protocols successfully proven in small animal models e.g. costimulatory blockade, T cell depletion failed in patients. Consequently, there is a need for more predictive transplant models to evaluate novel promising strategies, such as adoptive transfer of regulatory T cells (Treg). We established a clinically more relevant, life-supporting rat kidney transplant model using a high responder (DA to LEW) recipients that received donor-specific CD4(+)/ 8(+) GFP(+) T(mem) before transplantation to achieve similar pre-transplant frequencies of donor-specific T(mem) as seen in many patients. T cell depletion alone induced long-term graft survival in naïve recipients but could not prevent acute rejection in T(mem)(+) rats, like in patients. Only if T cell depletion was combined with permanent CNI-treatment, the intragraft inflammation, and acute/chronic allograft rejection could be controlled long-term. Remarkably, combining 10 days CNI treatment and adoptive transfer of Tregs (day 3) but not Treg alone also induced long-term graft survival and an intragraft tolerance profile (e.g. high TOAG-1) in T(mem)(+) rats. Our model allows evaluation of novel therapies under clinically relevant conditions.


Subject(s)
Calcineurin Inhibitors , Graft Rejection , Immunosuppressive Agents/pharmacology , Kidney Transplantation , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Flow Cytometry , Immunologic Memory , Lymphocyte Depletion , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction
5.
Diabetologia ; 53(10): 2105-11, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20593160

ABSTRACT

Significant progress has been made in recent years in the characterisation of the signal pathways of beta cell dysfunction and death in the pathogenesis of type 2 diabetes. Glucolipotoxicity acts as an exogenous factor whereas oxidative stress and endoplasmic reticulum stress may result from the processes of signal recognition and stimulated secretion within the beta cell. The pharmacological stimulation of secretion may thus appear to be a double-edged sword: it counteracts hyperglycaemia, but may do so at the expense of beta cell mass. So, in the long run, insulinotropic glucose-lowering drugs might do more harm than good. However, much of this logic is derived by analogy from the long-held assumption that beta cell hypersecretion imposed by insulin resistance causes the absolute secretion deficit in the later course of type 2 diabetes. In this concept the beta cell has a secondary role and loss of beta cell mass is necessary for the manifestation of type 2 diabetes. Recent studies have shown that a secretion deficit can exist well before insulin resistance and that major genetic risk factors concern beta cell function. Also, the evidence for a beta cell toxic effect of insulinotropic drugs is currently inconclusive. Assuming that the insulin secretion deficit is of pathogenetic importance in a network with insulin resistance as an aggravating factor, an insulinotropic glucose-lowering drug may do more good than harm if it relieves the beta cell from the stress of glucose overstimulation and does so without inducing hypoglycaemia.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents/adverse effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects
6.
Cell Death Dis ; 1: e23, 2010.
Article in English | MEDLINE | ID: mdl-21364631

ABSTRACT

Galectin-1 (gal-1), an endogenous ß-galactoside-binding protein, triggers T-cell death through several mechanisms including the death receptor and the mitochondrial apoptotic pathway. In this study we first show that gal-1 initiates the activation of c-Jun N-terminal kinase (JNK), mitogen-activated protein kinase kinase 4 (MKK4), and MKK7 as upstream JNK activators in Jurkat T cells. Inhibition of JNK activation with sphingomyelinase inhibitors (20 µM desipramine, 20 µM imipramine), with the protein kinase C-δ (PKCδ) inhibitor rottlerin (10 µM), and with the specific PKCθ pseudosubstrate inhibitor (30 µM) indicates that ceramide and phosphorylation by PKCδ and PKCθ mediate gal-1-induced JNK activation. Downstream of JNK, we observed increased phosphorylation of c-Jun, enhanced activating protein-1 (AP-1) luciferase reporter, and AP-1/DNA-binding in response to gal-1. The pivotal role of the JNK/c-Jun/AP-1 pathway for gal-1-induced apoptosis was documented by reduction of DNA fragmentation after inhibition JNK by SP600125 (20 µM) or inhibition of AP-1 activation by curcumin (2 µM). Gal-1 failed to induce AP-1 activation and DNA fragmentation in CD3-deficient Jurkat 31-13 cells. In Jurkat E6.1 cells gal-1 induced a proapoptotic signal pattern as indicated by decreased antiapoptotic Bcl-2 expression, induction of proapoptotic Bad, and increased Bcl-2 phosphorylation. The results provide evidence that the JNK/c-Jun/AP-1 pathway plays a key role for T-cell death regulation in response to gal-1 stimulation.Cell Death and Disease (2010) 1, e23; doi:10.1038/cddis.2010.1; published online 4 February 2010.


Subject(s)
Galectin 1/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Transcription Factor AP-1/metabolism , Anthracenes/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Death/drug effects , DNA/metabolism , Enzyme Activation/drug effects , Genes, Reporter , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Jurkat Cells , Kinetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , T-Lymphocytes/enzymology , bcl-Associated Death Protein/metabolism
7.
Diabetologia ; 52(7): 1381-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19367386

ABSTRACT

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.


Subject(s)
Adoptive Transfer/methods , CD8-Positive T-Lymphocytes/transplantation , Diabetes Mellitus, Type 1/prevention & control , Lymph Nodes/immunology , Pancreas/immunology , Prediabetic State/therapy , Animals , Blood Glucose , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , Cytokines/genetics , Cytokines/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Gene Expression/immunology , Immunophenotyping , Prediabetic State/immunology , Rats , Rats, Inbred Lew , Rats, Nude , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
8.
Cell Transplant ; 16(5): 527-37, 2007.
Article in English | MEDLINE | ID: mdl-17708342

ABSTRACT

Islet transplantation is a promising treatment in type 1 diabetes, but the need for chronic immunosuppression is a major hurdle to broad applicability. Ex vivo introduction of agents by lentiviral vectors-improving beta-cell resistance against immune attack-is an attractive path to pursue. The aim of this study was to investigate whether dissociation of islets to single cells prior to viral infection and reaggregation before transplantation would improve viral transduction efficacy without cytotoxicity. This procedure improved transduction efficacy with a LV-pWPT-CMV-EGFP construct from 11.2 +/- 4.1% at MOI 50 in whole islets to 80.0 +/- 2.8% at MOI 5. Viability (as measured by Hoechst/PI) and functionality (as measured by glucose challenge) remained high. After transplantation, the transfected pseudoislet aggregates remained EGFP positive for more than 90 days and the expression of EGFP colocalized primarily with the insulin-positive beta-cells. No increased vulnerability to immune attack was observed in vitro or in vivo. These data demonstrate that dispersion of islets prior to lentiviral transfection and reaggregation prior to transplantation is a highly efficient way to introduce genes of interest into islets for transplantation purposes in vitro and in vivo, but the amount of beta-cells needed for normalization of glycemia was more than eightfold higher when using dispersed cell aggregates versus unmanipulated islets. The high price to pay to reach stable and strong transgene expression in islet cells is certainly an important cell loss.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Lentivirus/genetics , Transduction, Genetic , Animals , Cell Aggregation/drug effects , Cell Death/drug effects , Cell Line , Cell Separation , Cell Survival/drug effects , Cytokines/pharmacology , Flow Cytometry , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Rats , Rats, Wistar , Time Factors , Transgenes
9.
Dtsch Med Wochenschr ; 131 Suppl 8: S226-30, 2006 Dec.
Article in German | MEDLINE | ID: mdl-17139575

ABSTRACT

Using the metabolic stimulus-secretion coupling of pancreatic beta cells as an example, this review illustrates how new strategies in the treatment of type 2 diabetes mellitus can be developed from the results of basic research. Metabolic stimulus-secretion coupling presupposes the metabolizing of those stimuli of insulin secretion that have the properties of nutritional substances. Changes in the ATP/ADP ratio within the beta cells will then trigger the release of insulin granules from them. Glucokinase, a glucose phosphorylating enzyme, functions as a metabolic glucose sensor, which couples changes in physiological glucose concentration in the pancreatic beta cells and in the liver to the intermediary metabolism, i.e. glycolysis, the citrate cycle and respiratory-chain phosphorylation. In this way insulin secretion and hepatic metabolism are positively influenced. Several pharmaceutical companies (Roche, Merck, Astra-Zeneca, Lilly) have recently developed first examples of glucokinase-activating compounds and demonstrated in animal models their efficacy in the treatment of type 2 diabetes mellitus. These glucokinase activators prevent glucokinase from changing into a catalytically inactive structure. They also increase glucose affinity of the enzyme and stabilize a catalytically active form of glucokinase proteins. In this way glucokinase activators increase glucose-induced insulin secretion and inhibit hepatic glucogenesis. Glucokinase activators are an interesting innovation in the future treatment of type 2 diabetes, because their action on beta cells and the liver is caused by changes in blood glucose concentration.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/therapy , Glucokinase/metabolism , Insulin-Secreting Cells/physiology , Insulin/metabolism , Liver/metabolism , Animals , Blood Glucose/physiology , Diabetes Mellitus, Type 2/metabolism , Glucokinase/drug effects , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Oxidative Phosphorylation , Pyridines/therapeutic use
10.
Diabetologia ; 48(8): 1541-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15986238

ABSTRACT

AIMS/HYPOTHESIS: Free radicals generated in mitochondria play a crucial role in the toxic effects of cytokines upon insulin-producing cells. This study therefore investigated the role of manganese superoxide dismutase (MnSOD) in cytokine-mediated toxicity in insulin-producing cells. METHODS: MnSOD was either stably overexpressed (MnSODsense) or stably suppressed (MnSODantisense) in insulin-producing RINm5F cells. Cell viability was quantified after incubation with different chemical reactive oxygen species (ROS) generators and with cytokines (IL-1beta alone or a mixture of IL-1beta, TNF-alpha and IFN-gamma). Additionally, cell proliferation and endogenous MnSOD protein expression were determined after exposure to cytokines. RESULTS: After incubation with hydrogen peroxide (H(2)O(2)) or hypoxanthine/xanthine oxidase no significant differences were observed in viability between control and MnSODsense or MnSODantisense clones. MnSOD overexpression reduced the viability of MnSODsense cells after exposure to the intracellular ROS generator menadione compared with control and MnSODantisense cells. MnSODsense cells also showed the highest susceptibility to cytokine toxicity with more than 75% loss of viability and a significant reduction of the proliferation rate after 72 h of incubation with a cytokine mixture. In comparison with control cells (67% viability loss), the reduction of viability in MnSODantisense cells was lower (50%), indicating a sensitising role of MnSOD in the progression of cytokine toxicity. The cell proliferation rate decreased in parallel to the reduction of cell viability. The MnSOD expression level after exposure to cytokines was also significantly lower in MnSODantisense cells than in control or MnSODsense cells. CONCLUSIONS/INTERPRETATION: The increase of the mitochondrial imbalance between the superoxide- and the H(2)O(2)-inactivating enzyme activities corresponds with a greater susceptibility to cytokines. Thus optimal antioxidative strategies to protect insulin-producing cells against cytokine toxicity may comprise a combined overexpression of H(2)O(2)-inactivating enzymes or suppression of MnSOD activity.


Subject(s)
Cytokines/toxicity , Insulin/biosynthesis , Mitochondria/enzymology , Reactive Oxygen Species/toxicity , Superoxide Dismutase/physiology , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , Hydrogen Peroxide/pharmacology , Interferon-gamma/toxicity , Interleukin-1/toxicity , Oxidants/pharmacology , RNA, Antisense/biosynthesis , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Tetrazolium Salts , Thiazoles , Tumor Necrosis Factor-alpha/toxicity , Vitamin K 3/pharmacology
12.
Diabetologia ; 47(7): 1292-1302, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15248046

ABSTRACT

AIMS/HYPOTHESIS: Cytokines are important humoral mediators of beta cell destruction in autoimmune diabetes. The aim of this study was to identify novel cytokine-induced genes in insulin-producing RINm5F cells, which may contribute to beta cell death or survival. METHODS: A global gene expression profile in cytokine-exposed insulin-producing RINm5F cells was achieved by automated restriction fragment differential display PCR. The expression of selected candidate genes was confirmed by real-time RT-PCR analysis. RESULTS: Exposure of RINm5F cells to IL-1beta or to a cytokine mixture (IL-1beta, TNF-alpha, IFN-gamma) for 6 h resulted in the differential expression of a functional gene cluster. Apart from the well-known up-regulation of the cytokine-responsive genes iNOS, NF-kappaB, MnSOD and Hsp70, several genes that belong to the functional cluster of the endocytotic pathway were identified. These endocytotic genes comprised: clathrin, megalin, synaptotagmin and calcineurin, which were up-regulated by IL-1beta or the cytokine mixture. In contrast, the expression of the calcineurin inhibitor CAIN and of the GDP/GTP exchange protein Rab3 was down-regulated by cytokines. Other up-regulated cytokine-responsive genes were: agrin, murine adherent macrophage protein mRNA ( MAMA) and transport-associated protein ( TAP1/MTP), whereas the plasma membrane calcium ATPase ( PMCA) 2 and PMCA 3 genes were down-regulated by cytokines. CONCLUSIONS/INTERPRETATION: Our results indicate that genes of the endocytotic pathway are regulated by pro-inflammatory cytokines. This might affect the density of cytokine receptors at the beta cell surface and concomitantly the sensitivity of the cells to cytokine toxicity. A better understanding of the functional cross-talk between endocytotic and cytokine signalling pathways could further the development of novel strategies to protect pancreatic beta cells against toxic effects of pro-inflammatory cytokines.


Subject(s)
Cytokines/immunology , Gene Expression Regulation, Neoplastic/immunology , Insulin/metabolism , Agrin/genetics , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Insulin Secretion , Insulinoma , Neoplasm Proteins/genetics , Pancreatic Neoplasms , RNA, Messenger/genetics , Rats
13.
J Mol Endocrinol ; 31(3): 509-18, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14664711

ABSTRACT

Pancreatic beta-cells have low activities of the antioxidant enzyme catalase. Nitric oxide interacts with the haem group of catalase inhibiting its activity. We have studied the activity of catalase in beta-cells under conditions mimicking prediabetes and in which nitric oxide is generated from cytokine treatment in vitro. We also studied whether there is regulation of catalase enzyme activity by nitric oxide at the protein or gene expression level. RINm5F insulin-producing cells, treated for 24 h with cytokines, showed increased medium nitrite production (17+/-2.2 vs 0.3+/-0.2 pmol/ micro g protein) and significantly decreased cellular catalase activity (42.4+/-4.5%) compared with control cells. A similar reduction was seen in catalase-overexpressing RIN-CAT cells and in rat or human pancreatic islets of Langerhans. Catalase activity was also suppressed by the long-acting nitric oxide donor diethylenetriamine/nitric oxide adduct (Deta-NO) and this inhibition was reversible. The inhibition of catalase activity by cytokines in RINm5F cells was significantly reversed by the addition of the nitric oxide synthase 2 (NOS2) inhibitors nitro monomethylarginine or N-(3-(aminomethyl)benzyl)acetamidine (1400W). Protein expression was found to be unchanged in cytokine- or Deta-NO-treated RINm5F cells, while mRNA expression was marginally increased. We have shown that inhibition of catalase activity by cytokines is nitric oxide dependent and propose that this inhibition may confer increased susceptibility to cytokine- or nitric oxide-induced cell killing.


Subject(s)
Catalase/metabolism , Cytokines/pharmacology , Insulinoma/enzymology , Nitric Oxide/pharmacology , Pancreatic Neoplasms/enzymology , Amidines/pharmacology , Animals , Benzylamines/pharmacology , Cell Death/physiology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , Insulin/metabolism , Islets of Langerhans/enzymology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/genetics , Rats , Triazenes/pharmacology , Tumor Cells, Cultured
15.
Diabetologia ; 45(11): 1542-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12436338

ABSTRACT

AIMS/HYPOTHESIS: We investigated the importance of the low affinity GLUT2 glucose transporter in the diabetogenic action of alloxan in bioengineered RINm5F insulin-producing cells with different expressions of the transporter. METHODS: GLUT2 glucose transporter expressing RINm5F cells were generated through stable transfection of the rat GLUT2 cDNA under the control of the cytomegalovirus promoter in the pcDNA3 vector. Viability of the cells was determined using a microtitre plate-based 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) assay. RESULTS: Cells expressing the GLUT2 transporter were susceptible to alloxan toxicity due to the uptake of alloxan by this specific glucose transporter isoform. The extent of the toxicity of alloxan was dependent upon the GLUT2 protein expression in the cells. The lipophilic alloxan derivative, butylalloxan, was toxic also to non-transfected control cells. Expression of the GLUT2 glucose transporter caused only a marginal increase in the toxicity of this substance. Butylalloxan, unlike alloxan itself, is not diabetogenic in vivo although, like the latter substance, it is beta-cell toxic in vitro through its ability to generate free radicals during redox cycling with glutathione. CONCLUSION/INTERPRETATION: Our results are consistent with the central importance of selective uptake of alloxan through the low affinity GLUT2 glucose transporter for the pancreatic beta-cell toxicity and diabetogenicity of this substance. Redox cycling and the subsequent generation of oxygen free radicals leads to necrosis of pancreatic beta cells and thus to a state of insulin-dependent diabetes mellitus, well-known as alloxan diabetes in experimental diabetes research.


Subject(s)
Alloxan/toxicity , Glucose/pharmacology , Islets of Langerhans/pathology , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , 3-O-Methylglucose/pharmacology , Alloxan/analogs & derivatives , Alloxan/antagonists & inhibitors , Animals , Cells, Cultured , Gene Expression Regulation/physiology , Glucose Transporter Type 2 , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Microsomes/metabolism , Rats , Recombinant Proteins/metabolism
16.
Diabetologia ; 45(6): 851-5, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12107729

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Islets of Langerhans/physiology , Thyroxine/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Cell Division/drug effects , In Situ Nick-End Labeling , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Rats
17.
Diabetologia ; 44(9): 1189-96, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11596676

ABSTRACT

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.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Animals , Apoptosis , B-Lymphocytes/pathology , Blood Glucose/analysis , Diabetes Mellitus, Type 1/metabolism , Glycosuria , Hyperglycemia , Insulin/blood , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Ketone Bodies/urine , Killer Cells, Natural/pathology , Macrophages/pathology , Male , Microscopy, Electron , Polyuria , Rats , Rats, Inbred Lew , Rats, Mutant Strains , T-Lymphocytes/pathology
18.
Pancreas ; 23(2): 186-96, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11484921

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/pathology , Islets of Langerhans/pathology , Pancreas Transplantation/pathology , Animals , Blood Glucose/metabolism , Cell Division , Diabetes Mellitus, Experimental/blood , Glucokinase/metabolism , Glucose Transporter Type 2 , Immunohistochemistry , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Monosaccharide Transport Proteins/metabolism , Rats , Rats, Inbred Lew , Time Factors
19.
J Biol Chem ; 276(47): 43915-23, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11522786

ABSTRACT

The low affinity glucose-phosphorylating enzyme glucokinase shows the phenomenon of intracellular translocation in beta cells of the pancreas and the liver. To identify potential binding partners of glucokinase by a systematic strategy, human beta cell glucokinase was screened by a 12-mer random peptide library displayed by the M13 phage. This panning procedure revealed two consensus motifs with a high binding affinity for glucokinase. The first consensus motif, LSAXXVAG, corresponded to the glucokinase regulatory protein of the liver. The second consensus motif, SLKVWT, showed a complete homology to the bifunctional enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2), which acts as a key regulator of glucose metabolism. Through yeast two-hybrid analysis it became evident that the binding of glucokinase to PFK-2/FBPase-2 is conferred by the bisphosphatase domain, whereas the kinase domain is responsible for dimerization. 5'-Rapid amplification of cDNA ends analysis and Northern blot analysis revealed that rat pancreatic islets express the brain isoform of PFK-2/FBPase-2. A minor portion of the islet PFK-2/FBPase-2 cDNA clones comprised a novel splice variant with 8 additional amino acids in the kinase domain. The binding of the islet/brain PFK-2/ FBPase-2 isoform to glucokinase was comparable with that of the liver isoform. The interaction between glucokinase and PFK-2/FBPase-2 may provide the rationale for recent observations of a fructose-2,6-bisphosphate level-dependent partial channeling of glycolytic intermediates between glucokinase and glycolytic enzymes. In pancreatic beta cells this interaction may have a regulatory function for the metabolic stimulus-secretion coupling. Changes in fructose-2,6-bisphosphate levels and modulation of PFK-2/FBPase-2 activities may participate in the physiological regulation of glucokinase-mediated glucose-induced insulin secretion.


Subject(s)
Bacteriophage M13/genetics , Epitopes/chemistry , Peptide Library , Phosphofructokinase-2/metabolism , Receptors, Cytoplasmic and Nuclear/immunology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , Epitopes/genetics , Epitopes/metabolism , Islets of Langerhans/enzymology , Molecular Sequence Data , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Two-Hybrid System Techniques
20.
Biochim Biophys Acta ; 1523(1): 65-72, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-11099859

ABSTRACT

Glucose induces slow oscillations of the cytoplasmic Ca2+ concentration in pancreatic beta-cells. In order to elucidate the mechanisms responsible for the slow [Ca2+]i oscillations the effects of various nutrient insulin secretagogues on glucose-induced [Ca2+]i oscillations in intact mouse pancreatic islets and single beta-cells were studied. These were the glycolytic intermediates, glyceraldehyde and pyruvate, and the mitochondrial substrate, alpha-ketoisocaproic acid (KIC). Glucose, at a 10 or 15 mM concentration, induced the typical slow oscillations of [Ca2+]i (0.4 min(-1)). At higher glucose concentrations the frequency of these oscillations decreased further (0.2 min(-1)). Glyceraldehyde, an insulin secretagogue like glucose, did not cause slow oscillations of [Ca2+]i in the absence of glucose. However, it exhibited a synergistic action with glucose. Glyceraldehyde, at 3 or 10 mM concentration, induced slow [Ca2+]i oscillations at a substimulatory concentration of 5 mM glucose (0.3-0.4 min(-1)) and reduced the frequency of the glucose-induced [Ca2+]i oscillations at stimulatory concentrations of 10 or 15 mM glucose (0.2 min(-1)). KIC (5 or 10 mM) as well as pyruvate (10 mM), the end product of glycolysis, and its ester methyl pyruvate (10 mM), did not cause slow oscillations of [Ca2+]i in the absence of glucose. In contrast to glyceraldehyde, however, all three compounds were capable of preventing the slow [Ca2+]i oscillations induced by glucose. Mannoheptulose (2 mM), an inhibitor of glucokinase and glucose-induced insulin secretion, reversibly blocked any kind of [Ca2+]i oscillation and returned the [Ca2+]i to a basal level through its ability to inhibit glycolytic flux. It can be concluded therefore that only substrates which generate a glucokinase-mediated metabolic flux through glycolysis and produce glycolytic ATP can induce slow [Ca2+]i oscillations in pancreatic beta-cells.


Subject(s)
Calcium/metabolism , Glucose/pharmacology , Glyceraldehyde/pharmacology , Glycolysis , Islets of Langerhans/physiology , Keto Acids/pharmacology , Animals , Cells, Cultured , Female , Islets of Langerhans/drug effects , Kinetics , Mice , Mice, Inbred Strains , Oscillometry , Pyruvates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL