Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Diabetol ; 21(1): 5, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34991588

ABSTRACT

BACKGROUND: Systemic inflammatory processes plausibly contribute to the development of cardiovascular complications, causing increased morbidity and mortality in type 2 diabetes. Circulating inflammatory markers, i.e., interleukin (IL)-6 and tumour necrosis factor-α, are associated with neurocardiac measures. We examined a broad panel of various inflammatory and inflammation-related serum markers to obtain more detailed insight into the possible neuro-immune interaction between cardiovascular regulation and systemic level of inflammation. METHODS: Serum samples from 100 participants with type 2 diabetes were analysed. Heart rate variability, cardiovascular autonomic reflex tests, and cardiac vagal tone tests were performed based on electrocardiographic readings. Data regarding covariates (demographic-, diabetes-, and cardiovascular risk factors) were registered. RESULTS: Increased serum levels of IL-12/IL-23p40 (p < 0.01) and intercellular adhesion molecule (ICAM)-1 (p < 0.007) were associated with diminished heart rate variability measures. After all adjustments, the associations between IL-12/23p40, SDANN and VLF persisted (p = 0.001). Additionally, serum levels of vascular endothelial growth factor (VEGF)-C were associated with response to standing (p = 0.005). DISCUSSION: The few but robust associations between neurocardiac regulation and serum markers found in this study suggest systemic changes in proinflammatory, endothelial, and lymphatic function, which collectively impacts the systemic cardiovascular function. Our results warrant further exploration of IL-12/IL-23p40, ICAM-1, and VEGF-C as possible cardiovascular biomarkers in T2D that may support future decisions regarding treatment strategies for improved patient care.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/blood , Heart Rate , Inflammation Mediators/blood , Interleukin-12 Subunit p40/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Electrocardiography , Female , Humans , Intercellular Adhesion Molecule-1/blood , Male , Middle Aged , Predictive Value of Tests , Prognosis , Up-Regulation , Vascular Endothelial Growth Factor C/blood
2.
Adv Clin Chem ; 75: 99-158, 2016.
Article in English | MEDLINE | ID: mdl-27346618

ABSTRACT

The discovery 30 years ago that inflammatory cytokines cause a concentration, activity, and time-dependent bimodal response in pancreatic ß-cell function and viability has been a game-changer in the fields of research directed at understanding inflammatory regulation of ß-cell function and survival and the causes of ß-cell failure and destruction in diabetes. Having until then been confined to the use of pathophysiologically irrelevant ß-cell toxic chemicals as a model of ß-cell death, researchers could now mimic endocrine and paracrine effects of the cytokine response in vitro by titrating concentrations in the low to the high picomolar-femtomolar range and vary exposure time for up to 14-16h to reproduce the acute regulatory effects of systemic inflammation on ß-cell secretory responses, with a shift to inhibition at high picomolar concentrations or more than 16h of exposure to illustrate adverse effects of local, chronic islet inflammation. Since then, numerous studies have clarified how these bimodal responses depend on discrete signaling pathways. Most interest has been devoted to the proapoptotic response dependent upon mainly nuclear factor κ B and mitogen-activated protein kinase activation, leading to gene expressional changes, endoplasmic reticulum stress, and triggering of mitochondrial dysfunction. Preclinical studies have shown preventive effects of cytokine antagonism in animal models of diabetes, and clinical trials demonstrating proof of concept are emerging. The full clinical potential of anticytokine therapies has yet to be shown by testing the incremental effects of appropriate dosing, timing, and combinations of treatments. Due to the considerable translational importance of enhancing the precision, specificity, and safety of antiinflammatory treatments of diabetes, we review here the cellular, preclinical, and clinical evidence of which of the death pathways recently proposed in the Nomenclature Committee on Cell Death 2012 Recommendations are activated by inflammatory cytokines in the pancreatic ß-cell to guide the identification of antidiabetic targets. Although there are still scarce human data, the cellular and preclinical studies point to the caspase-dependent intrinsic apoptosis pathway as the prime effector of inflammatory ß-cell apoptosis.


Subject(s)
Apoptosis , Cytokines/metabolism , Insulin-Secreting Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cytokines/genetics , Cytokines/pharmacology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Endoplasmic Reticulum Stress , Humans
3.
Cell Mol Life Sci ; 65(7-8): 1248-55, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18278435

ABSTRACT

An insufficient number of insulin-producing beta-cells is a major cause of defective control of blood glucose in both type 1 and type 2 diabetes. The aim of this study was to clarify whether the insulinotropic imidazolines can affect the survival of highly proliferating insulin-secreting cells, here exemplified by the MIN6 cell line. Our data demonstrate that RX871024, but not efaroxan, triggered MIN6 cell death and potentiated death induced by a combination of the pro-inflammatory cytokines interleukin-1beta, interferon- gamma and tumor necrosis factor-alpha. These effects did not involve changes in nitric oxide production but correlated with stimulation of c-jun N-terminal kinase (JNK) activity and activation of caspases-1, -3, -8 and -9. Our results suggest that the imidazoline RX871024 causes death of highly proliferating insulin-secreting cells, putatively via augmentation of JNK activity, a finding that may impact on the possibility of using compounds of similar activity in the treatment of diabetes.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Benzofurans/pharmacology , Caspases/metabolism , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cytokines/pharmacology , Enzyme Activation/drug effects , Humans , Insulin-Secreting Cells/drug effects , Mice , Nitric Oxide/biosynthesis
4.
Diabetologia ; 50(4): 779-89, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17265033

ABSTRACT

AIMS/HYPOTHESIS: The immune-mediated elimination of pancreatic beta cells in type 1 diabetes involves release of cytotoxic cytokines such as IL-1beta and IFNgamma, which induce beta cell death in vitro by mechanisms that are both dependent and independent of nitric oxide (NO). Nuclear factor kappa B (NFkappaB) is a critical signalling molecule in inflammation and is required for expression of the gene encoding inducible NO synthase (iNOS) and of pro-apoptotic genes. NFkappaB has recently been shown to associate with chromatin-modifying enzymes histone acetyltransferases and histone deacetylases (HDAC), and positive effects of HDAC inhibition have been obtained in several inflammatory diseases. Thus, the aim of this study was to investigate whether HDAC inhibition protects beta cells against cytokine-induced toxicity. MATERIALS AND METHODS: The beta cell line, INS-1, or intact rat islets were precultured with HDAC inhibitors suberoylanilide hydroxamic acid or trichostatin A in the absence or presence of IL-1beta and IFNgamma. Effects on insulin secretion and NO formation were measured by ELISA and Griess reagent, respectively. iNOS levels and NFkappaB activity were measured by immunoblotting and by immunoblotting combined with electrophoretic mobility shift assay, respectively. Viability was analysed by 3-(4,5-dimethyldiazol-2-yl)-2,5-diphenyl-tetrazolium bromide and apoptosis by terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) assay and histone-DNA complex ELISA. RESULTS: HDAC inhibition reduced cytokine-mediated decrease in insulin secretion and increase in iNOS levels, NO formation and apoptosis. IL-1beta induced a bi-phasic phosphorylation of inhibitor protein kappa Balpha (IkappaBalpha) with the 2nd peak being sensitive to HDAC inhibition. No effect was seen on IkappaBalpha degradation and NFkappaB DNA binding. CONCLUSIONS/INTERPRETATION: HDAC inhibition prevents cytokine-induced beta cell apoptosis and impaired beta cell function associated with a downregulation of NFkappaB transactivating activity.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation, Enzymologic , Histone Deacetylase Inhibitors , Insulin-Secreting Cells/metabolism , Animals , Apoptosis , Cell Survival , Enzyme Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Recombinant Proteins/chemistry , Signal Transduction
5.
Diabetologia ; 49(8): 1864-75, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16718462

ABSTRACT

AIMS/HYPOTHESIS: IL-1beta released from immune cells induces beta cell pro-apoptotic signalling via mitogen-activated protein kinases (MAPKs) and nuclear factor-kappaB (NF-kappaB). In neurons, the neural cell adhesion molecule (NCAM) signals to several elements involved in IL-1beta-induced pro-apoptotic signalling in beta cells. Pancreatic beta cells express NCAM, but its biological effects in these cells are unclear. The aim of this study was to investigate whether there is cross-talk between NCAM signalling and cytokine-induced pro-apoptotic signalling. MATERIALS AND METHODS: Western blotting was used to investigate levels of NCAM and inducible nitric oxide synthase, phosphorylation of Src and MAPKs, and cleavage of caspase-3. MAPK activity was investigated with an in vitro kinase assay. Apoptosis was detected by cleaved caspase-3 and a Cell Death Detection ELISA(plus) assay. NCAM-induced fibroblast growth factor receptor (FGFR) activation was investigated in NCAM(-/-) Trex293 cells where FGFR phosphorylation was measured by Western blotting after NCAM transfection. RESULTS: Pre-exposure of INS-1E cells to the FGFR-inhibitor SU5402, but not to the Src-inhibitor PP2, dose-dependently inhibited IL-1beta-mediated MAPK activity. A synthetic peptide, C3d, reported to bind NCAM, did not activate MAPK or Akt as reported in neurons but inhibited IL-1beta-induced MAPK activity, thereby mimicking the effect of SU5402. Furthermore, C3d inhibited NCAM-induced FGFR phosphorylation and apoptosis induced by IL-1beta plus IFN-gamma, but did not affect IL-1beta-induced NF-kappaB signalling. CONCLUSIONS/INTERPRETATION: We suggest that NCAM signalling through FGFR is required for efficient IL-1beta pro-apoptotic signalling by facilitating IL-1beta-induced MAPK activation downstream of the NF-kappaB-MAPK branching point. Further, these data identify a novel function of C3d as an inhibitor of NCAM-induced FGFR activity and of IL-1beta-induced MAPK activation in beta cells.


Subject(s)
Insulin-Secreting Cells/physiology , Interleukin-1/pharmacology , Neural Cell Adhesion Molecules/physiology , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction/drug effects , Animals , Cell Line , Complement C3d/physiology , Hippocampus/physiology , Insulin-Secreting Cells/drug effects , Insulinoma , Neurons/drug effects , Neurons/physiology , Pancreatic Neoplasms , Phosphorylation , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
6.
Diabetologia ; 48(12): 2582-90, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16283237

ABSTRACT

AIMS/HYPOTHESIS: The beta cell destruction and insulin deficiency that characterises type 1 diabetes mellitus is partially mediated by cytokines, such as IL-1beta, and by nitric oxide (NO)-dependent and -independent effector mechanisms. IL-1beta activates mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase (ERK), p38 and c-Jun NH2-terminal kinase (JNK), and the nuclear factor kappa B (NFkappaB) pathway. Both pathways are required for expression of the gene encoding inducible nitric oxide synthase (iNOS) and for IL-1beta-mediated beta cell death. The molecular mechanisms by which these two pathways regulate beta cell Nos2 expression are currently unknown. Therefore, the aim of this study was to clarify the putative crosstalk between MAPK and NFkappaB activation in beta cells. MATERIALS AND METHODS: The MAPKs ERK, p38 and JNK were inhibited by SB203580, PD98059 or Tat-JNK binding domain or by cells overexpressing the JNK binding domain. The effects of MAPK inhibition on IL-1beta-induced iNOS production and kappa B inhibitor protein (IkappaB) degradation were examined by western blotting. NFkappaB DNA binding was investigated by electrophoretic mobility shift assay, while NFkappaB-induced gene transcription was evaluated by gene reporter assays. RESULTS: Inhibition of the MAPKs did not affect IkappaB degradation or NFkappaB DNA binding. However, inhibition of ERK reduced NFkappaB-mediated Nos2 expression; serine 276 phosphorylation of the p65 unit of the NFkappaB complex seemed critical, as evaluated by amino acid mutation analysis. CONCLUSIONS/INTERPRETATION: ERK activity is required for NFkappaB-mediated transcription of Nos2 in insulin-producing INS-1E cells, indicating that ERK regulates Nos2 expression by increasing the transactivating capacity of NFkappaB. This may involve phosphorylation of Ser276 on p65 by an as yet unidentified kinase.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Gene Expression , Insulin-Secreting Cells/physiology , Insulin/metabolism , Interleukin-1/physiology , NF-kappa B/physiology , Nitric Oxide Synthase Type II/genetics , Animals , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Humans , Imidazoles/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Interleukin-1/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/physiology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/physiology , Nitric Oxide Synthase Type II/biosynthesis , Phosphorylation , Pyridines/pharmacology , Rats , Serine/analysis , Synaptotagmin I/chemistry , Synaptotagmin I/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/physiology
7.
Diabetologia ; 48(10): 2039-50, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16132952

ABSTRACT

AIMS/HYPOTHESIS: Pro-inflammatory cytokines cause beta cell secretory dysfunction and apoptosis--a process implicated in the pathogenesis of type 1 diabetes. Cytokines induce the expression of inducible nitric oxide (NO) synthase (iNOS) leading to NO production. NO contributes to cytokine-induced apoptosis, but the underlying mechanisms are unclear. The aim of this study was to investigate whether NO modulates signalling via mitogen-activated protein kinases (MAPKs) and Akt. MATERIALS AND METHODS: MAPK activities in INS-1 cells and isolated islets were determined by immunoblotting and in vitro kinase assay. Apoptosis was determined by ELISA measurement of histone-DNA complexes present in cytoplasm. RESULTS: Apoptosis in INS-1 cells induced by IL-1beta plus IFNgamma was dependent on NO production as demonstrated by the use of the NOS blocker NG-methyl-L-arginine. Accordingly, an NO donor (S-nitroso-N-acetyl-D, L-penicillamine, SNAP) dose-dependently caused apoptosis in INS-1 cells. SNAP activated c-Jun N-terminal kinase (JNK) and p38 MAPK, but suppressed the activity of extracellular signal-regulated kinase MAPK. In rat islets, NOS inhibition decreased JNK and p38 activities induced by a 6-h exposure to IL-1beta. Likewise, IL-1beta-induced JNK and p38 activities were lower in iNOS(-/-) mouse islets than in wild-type islets. In human islets, SNAP potentiated IL-1beta-induced JNK activation. The constitutive level of active, Ser473-phosphorylated Akt in INS-1 cells was suppressed by SNAP. IGF-I activated Akt and protected against SNAP-induced apoptosis. The anti-apoptotic effect of IGF-I was not associated with reduced JNK activation. CONCLUSIONS/INTERPRETATION: We suggest that NO contributes to cytokine-induced apoptosis via potentiation of JNK activity and suppression of Akt.


Subject(s)
Apoptosis/drug effects , Cytokines/pharmacology , Insulin-Secreting Cells/drug effects , MAP Kinase Kinase 4/genetics , Nitric Oxide/physiology , Oncogene Protein v-akt/genetics , Animals , Blotting, Western , Cell Separation , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , S-Nitroso-N-Acetylpenicillamine/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...