Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Biomed Pharmacother ; 164: 114895, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37224758

ABSTRACT

BACKGROUND AND PURPOSE: Glucocorticoids (GCs) are the main treatment for autoimmune and inflammatory disorders and are also used as immunosuppressive therapy for patients with organ transplantation. However, these treatments have several side effects, including metabolic disorders. Indeed, cortico-therapy may induce insulin resistance, glucose intolerance, disrupted insulin and glucagon secretion, excessive gluconeogenesis, leading to diabetes in susceptible individuals. Recently, lithium has been shown to alleviate deleterious effects of GCs in various diseased conditions. EXPERIMENTAL APPROACH: In this study, using two rat models of GC-induced metabolic disorders, we investigated the effects of Lithium Chloride (LiCl) in the mitigation of deleterious effects of GCs. Rats were treated either with corticosterone or dexamethasone, and with or without LiCl. Animals were then assessed for glucose tolerance, insulin sensitivity, in vivo and ex vivo glucose-induced insulin secretion and hepatic gluconeogenesis. KEY RESULTS: We showed that in rats chronically treated with corticosterone, lithium treatment markedly reduced insulin resistance. In addition, in rats treated with dexamethasone, lithium administration improved glucose tolerance, associated with enhanced insulin secretion in vivo. Moreover, liver gluconeogenesis was reduced upon LiCl treatment. The improvement of insulin secretion in vivo appeared to be due to an indirect regulation of ß cell function, since the ex vivo assessment of insulin secretion and ß cell mass in islets from animals treated with LiCl revealed no difference compared to untreated animals. CONCLUSION AND IMPLICATIONS: Collectively, our data provide evidences for the beneficial effects of lithium to mitigate the adverse metabolic effects of chronic cortico-therapy.


Subject(s)
Diabetes Mellitus , Insulin Resistance , Rats , Animals , Insulin Resistance/physiology , Lithium/pharmacology , Corticosterone , Blood Glucose/metabolism , Glucocorticoids , Diabetes Mellitus/chemically induced , Insulin/metabolism , Glucose/metabolism , Gluconeogenesis , Dexamethasone/adverse effects , Lithium Compounds
2.
Chemosphere ; 314: 137691, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36592828

ABSTRACT

Since the use of bisphenol A (BPA) has been restricted because of its endocrine disruptor properties, bisphenol S (BPS) has been widely used as a substitute of BPA. However, BPS exerts similar effects on metabolic health as BPA. The effects of maternal exposure to BPA and BPS on the metabolic health of offspring have been largely documented during the past decade. However, the impact of preconceptional paternal exposure to BPS on progenies remains unexplored. In this study we investigated the impact of paternal exposure to BPS before conception, on the metabolic phenotype of offspring. Male Wistar rats were administered BPS through drinking water at the dose of 4 µg/kg/day (BPS-4 sires) or 40 µg/kg/day (BPS-40 sires) for 2 months before mating with females. The progenies (F1) were studied at fetal stage and in adulthood. We showed that preconceptional paternal exposure to BPS for 2 months did not alter the metabolic status of sires. The female offspring of sires exposed to lower or higher doses of BPS showed no alteration of their metabolic phenotype compared to females from control sires. In contrast, male offspring of BPS-4 sires exhibited increased body weight and body fat/lean ratio, decreased insulin sensitivity and increased glucose-induced insulin secretion at adult age, compared to the male offspring of control sires. Moreover, male offspring of BPS-4 sires developed glucose intolerance later in life. None of these effects were apparent in male offspring of BPS-40 sires. In conclusion, our study provides the first evidence of the non-monotonic and sex-specific effects of preconceptional paternal exposure to BPS on the metabolic health of offspring, suggesting that BPS is not a safe BPA substitute regarding the inter-generational transmission of metabolic disorders through the paternal lineage.


Subject(s)
Insulin Resistance , Prenatal Exposure Delayed Effects , Humans , Rats , Male , Female , Animals , Rats, Wistar , Maternal Exposure , Paternal Exposure/adverse effects , Glucose/metabolism , Benzhydryl Compounds/toxicity , Prenatal Exposure Delayed Effects/chemically induced
3.
Int J Mol Sci ; 23(23)2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36499613

ABSTRACT

The global prevalence of diabetes mellitus and Alzheimer's disease is increasing alarmingly with the aging of the population. Numerous epidemiological data suggest that there is a strong association between type 2 diabetes and an increased risk of dementia. These diseases are both degenerative and progressive and share common risk factors. The amyloid cascade plays a key role in the pathophysiology of Alzheimer's disease. The accumulation of amyloid beta peptides gradually leads to the hyperphosphorylation of tau proteins, which then form neurofibrillary tangles, resulting in neurodegeneration and cerebral atrophy. In Alzheimer's disease, apart from these processes, the alteration of glucose metabolism and insulin signaling in the brain seems to induce early neuronal loss and the impairment of synaptic plasticity, years before the clinical manifestation of the disease. The large amount of evidence on the existence of insulin resistance in the brain during Alzheimer's disease has led to the description of this disease as "type 3 diabetes". Available animal models have been valuable in the understanding of the relationships between type 2 diabetes and Alzheimer's disease, but to date, the mechanistical links are poorly understood. In this non-exhaustive review, we describe the main molecular mechanisms that may link these two diseases, with an emphasis on impaired insulin and IGF-1 signaling. We also focus on GSK3ß and DYRK1A, markers of Alzheimer's disease, which are also closely associated with pancreatic ß-cell dysfunction and type 2 diabetes, and thus may represent common therapeutic targets for both diseases.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Animals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Diabetes Mellitus, Type 2/metabolism , tau Proteins/metabolism , Insulin/metabolism , Brain/metabolism
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166509, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35914653

ABSTRACT

Type 2 diabetes is associated with an inflammatory phenotype in the pancreatic islets. We previously demonstrated that proinflammatory cytokines potently activate the tryptophan/kynurenine pathway (TKP) in INS-1 cells and in normal rat islets. Here we examined: (1) the TKP enzymes expression in the diabetic GK islets; (2) the TKP enzymes expression profiles in the GK islets before and after the onset of diabetes; (3) The glucose-stimulated insulin secretion (GSIS) in vitro in GK islets after KMO knockdown using specific morpholino-oligonucleotides against KMO or KMO blockade using the specific inhibitor Ro618048; (4) The glucose tolerance and GSIS after acute in vivo exposure to Ro618048 in GK rats. We report a remarkable induction of the kmo gene in GK islets and in human islets exposed to proinflammatory conditions. It occurred prominently in beta cells. The increased expression and activity of KMO reflected an acquired adaptation. Both KMO knockdown and specific inhibitor Ro618048 enhanced GSIS in vitro in GK islets. Moreover, acute administration of Ro618048 in vivo improved glucose tolerance, GSIS and basal blood glucose levels in GK rats. These results demonstrate that targeting islet TKP is able to correct defective GSIS. KMO inhibition could represent a potential therapeutic strategy for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Blood Glucose/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/metabolism , Morpholinos , Rats , Rats, Wistar , Tryptophan/metabolism
5.
Front Immunol ; 13: 896179, 2022.
Article in English | MEDLINE | ID: mdl-35677049

ABSTRACT

Type-2 diabetes is a complex disorder that is now considered to have an immune component, with functional impairments in many immune cell types. Type-2 diabetes is often accompanied by comorbid obesity, which is associated with low grade inflammation. However,the immune status in Type-2 diabetes independent of obesity remains unclear. Goto-Kakizaki rats are a non-obese Type-2 diabetes model. The limited evidence available suggests that Goto-Kakizaki rats have a pro-inflammatory immune profile in pancreatic islets. Here we present a detailed overview of the adult Goto-Kakizaki rat immune system. Three converging lines of evidence: fewer pro-inflammatory cells, lower levels of circulating pro-inflammatory cytokines, and a clear downregulation of pro-inflammatory signalling in liver, muscle and adipose tissues indicate a limited pro-inflammatory baseline immune profile outside the pancreas. As Type-2 diabetes is frequently associated with obesity and adipocyte-released inflammatory mediators, the pro-inflammatory milieu seems not due to Type-2 diabetes per se; although this overall reduction of immune markers suggests marked immune dysfunction in Goto-Kakizaki rats.


Subject(s)
Diabetes Mellitus, Type 2 , Animals , Biomarkers , Immune System , Obesity , Rats , Rats, Wistar
6.
J Cell Physiol ; 237(1): 881-896, 2022 01.
Article in English | MEDLINE | ID: mdl-34435368

ABSTRACT

Purinergic P2Y receptors, by binding adenosine triphosphate (ATP), are known for enhancing glucose-stimulated insulin secretion (GSIS) in pancreatic ß cells. However, the impact of these receptors in the actin dynamics and insulin granule exocytosis in these cells is not established, neither in normal nor in glucotoxic environment. In this study, we investigate the involvement of P2Y receptors on the behavior of insulin granules and the subcortical actin network dynamics in INS-1 832/13 ß cells exposed to normal or glucotoxic environment and their role in GSIS. Our results show that the activation of P2Y purinergic receptors by ATP or its agonist increase the insulin granules exocytosis and the reorganization of the subcortical actin network and participate in the potentiation of GSIS. In addition, their activation in INS-1832/13 ß-cells, with impaired insulin secretion following exposure to elevated glucose levels, restores GSIS competence through the distal steps of insulin exocytosis. These results are confirmed ex vivo by perifusion experiments on islets from type 2 diabetic (T2D) Goto-Kakizaki (GK) rats. Indeed, the P2Y receptor agonist restores the altered GSIS, which is normally lost in this T2D animal model. Moreover, we observed an improvement of the glucose tolerance, following the acute intraperitoneal injection of the P2Y agonist concomitantly with glucose, in diabetic GK rats. All these data provide new insights into the unprecedented therapeutic role of P2Y purinergic receptors in the pathophysiology of T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Actins/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Diabetes Mellitus, Type 2/metabolism , Exocytosis , Glucose/metabolism , Glucose/toxicity , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Rats , Receptors, Purinergic P2Y/metabolism
7.
Cell Death Dis ; 12(12): 1136, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876563

ABSTRACT

Glucocorticoids (GCs) are widely prescribed for their anti-inflammatory and immunosuppressive properties as a treatment for a variety of diseases. The use of GCs is associated with important side effects, including diabetogenic effects. However, the underlying mechanisms of GC-mediated diabetogenic effects in ß-cells are not well understood. In this study we investigated the role of glycogen synthase kinase 3 (GSK3) in the mediation of ß-cell death and dysfunction induced by GCs. Using genetic and pharmacological approaches we showed that GSK3 is involved in GC-induced ß-cell death and impaired insulin secretion. Further, we unraveled the underlying mechanisms of GC-GSK3 crosstalk. We showed that GSK3 is marginally implicated in the nuclear localization of GC receptor (GR) upon ligand binding. Furthermore, we showed that GSK3 regulates the expression of GR at mRNA and protein levels. Finally, we dissected the proper contribution of each GSK3 isoform and showed that GSK3ß isoform is sufficient to mediate the pro-apoptotic effects of GCs in ß-cells. Collectively, in this work we identified GSK3 as a viable target to mitigate GC deleterious effects in pancreatic ß-cells.


Subject(s)
Glucocorticoids , Glycogen Synthase Kinase 3 , Apoptosis , Cell Death , Glucocorticoids/adverse effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta/genetics
8.
Biomolecules ; 11(5)2021 05 18.
Article in English | MEDLINE | ID: mdl-34069853

ABSTRACT

The impact of maternal nutrition on offspring is well documented. However, the implication of pre-conceptional paternal nutrition on the metabolic health of the progeny remains underexplored. Here, we investigated the impact of paternal high-protein diet (HPD, 43.2% protein) consumption on the endocrine pancreas and the metabolic phenotype of offspring. Male Wistar rats were given HPD or standard diet (SD, 18.9% protein) for two months. The progenies (F1) were studied at fetal stage and in adulthood. Body weight, glycemia, glucose tolerance (GT), glucose-induced insulin secretion in vivo (GIIS) and whole-body insulin sensitivity were assessed in male and female F1 offspring. Insulin sensitivity, GT and GIIS were similar between F1 females from HPD (HPD/F1) and SD fathers (SD/F1). Conversely, male HPD/F1 exhibited increased insulin sensitivity (p < 0.05) and decreased GIIS (p < 0.05) compared to male SD/F1. The improvement of insulin sensitivity in HPD/F1 was sustained even after 2 months of high-fat feeding. In male HPD/F1, the ß cell mass was preserved and the ß cell plasticity, following metabolic challenge, was enhanced compared to SD/F1. In conclusion, we provide the first evidence of a sex-specific impact of paternal HPD on the insulin sensitivity and GIIS of their descendants, demonstrating that changes in paternal nutrition alter the metabolic status of their progeny in adulthood.


Subject(s)
Diet, High-Protein/adverse effects , Insulin Resistance , Insulin-Secreting Cells/metabolism , Paternal Exposure/adverse effects , Animals , Body Weight , Case-Control Studies , Female , Insulin-Secreting Cells/drug effects , Male , Rats , Rats, Wistar , Sex Characteristics
9.
Nat Commun ; 12(1): 1064, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594056

ABSTRACT

Polycystic ovary syndrome (PCOS) is characterized by an oligo-anovulation, hyperandrogenism and polycystic ovarian morphology combined with major metabolic disturbances. However, despite the high prevalence and the human and economic consequences of this syndrome, its etiology remains unknown. In this study, we show that female Goto-Kakizaki (GK) rats, a type 2 diabetes mellitus model, encapsulate naturally all the reproductive and metabolic hallmarks of lean women with PCOS at puberty and in adulthood. The analysis of their gestation and of their fetuses demonstrates that this PCOS-like phenotype is developmentally programmed. GK rats also develop features of ovarian hyperstimulation syndrome. Lastly, a comparison between GK rats and a cohort of women with PCOS reveals a similar reproductive signature. Thus, this spontaneous rodent model of PCOS represents an original tool for the identification of the mechanisms involved in its pathogenesis and for the development of novel strategies for its treatment.


Subject(s)
Polycystic Ovary Syndrome/pathology , Adiposity , Animals , Animals, Newborn , Body Weight , Discriminant Analysis , Disease Models, Animal , Dyslipidemias/pathology , Endocrine System/pathology , Estrous Cycle , Female , Glucose Tolerance Test , Gonadotropins/pharmacology , Hormones/blood , Humans , Insulin Secretion , Least-Squares Analysis , Lipids/chemistry , Male , Maternal-Fetal Exchange , Multivariate Analysis , Ovary/pathology , Ovary/physiopathology , Phenotype , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Rats, Wistar , Reproduction , Sexual Maturation
10.
J Endocrinol ; 244(1): 133-148, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31600727

ABSTRACT

Islet inflammation is associated with defective ß cell function and mass in type 2 diabetes (T2D). Glycogen synthase kinase 3 (GSK3) has been identified as an important regulator of inflammation in different diseased conditions. However, the role of GSK3 in islet inflammation in the context of diabetes remains unexplored. In this study, we investigated the direct implication of GSK3 in islet inflammation in vitro and tested the impact of GSK3 inhibition in vivo, on the reduction of islet inflammation, and the improvement of glucose metabolism in the Goto-Kakizaki (GK) rat, a spontaneous model of T2D. GK rats were chronically treated with infra-therapeutic doses of lithium, a widely used inhibitor of GSK3. We analyzed parameters of glucose homeostasis as well as islet inflammation and fibrosis in the endocrine pancreas. Ex vivo, we tested the impact of GSK3 inhibition on the autonomous inflammatory response of non-diabetic rat and human islets, exposed to a mix of pro-inflammatory cytokines to mimic an inflammatory environment. Treatment of young GK rats with lithium prevented the development of overt diabetes. Lithium treatment resulted in reduced expression of pro-inflammatory cytokines in the islets. It decreased islet fibrosis and partially restored the glucose-induced insulin secretion in GK rats. Studies in non-diabetic human and rat islets exposed to inflammatory environment revealed the direct implication of GSK3 in the islet autonomous inflammatory response. We show for the first time, the implication of GSK3 in islet inflammation and suggest this enzyme as a viable target to treat diabetes-associated inflammation.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glycogen Synthase Kinase 3/metabolism , Islets of Langerhans/metabolism , Animals , Disease Models, Animal , Fibrosis , Glucose/metabolism , Humans , Inflammation , Insulin Secretion , Male , Rats , Rats, Wistar
11.
Biochim Biophys Acta ; 1862(8): 1401-11, 2016 08.
Article in English | MEDLINE | ID: mdl-27101990

ABSTRACT

Actin dynamics in pancreatic ß-cells is involved in insulin exocytosis but the molecular mechanisms of this dynamics and its role in biphasic insulin secretion in pancreatic ß-cells is largely unknown. Moreover, the impact of a glucotoxic environment on the sub-cortical actin network dynamics is poorly studied. In this study, we investigate the behavior of insulin granules and the subcortical actin network dynamics in INS-1 832/13 ß-cells submitted to a normal or glucotoxic environment. Our results show that glucose stimulation leads to a reorganization of the subcortical actin network with a rupture of its interactions with t-SNARE proteins (Syntaxin 1A and SNAP-25), promoting insulin secretion in INS-1 832/13 ß-cells. Prolonged exposure of INS-1 832/13 ß-cells to high-glucose levels (glucotoxicity) leads to the densification of the cortical actin network, which prevents its reorganization under acute glucose, and diminishes the glucose-stimulated insulin secretion, as shown by the decreased number of fusion events. The most interesting in our results is the partial restoration by GLP-1 of the insulin secretion ability from high-glucose treated INS-1 832/13 cells. This improved insulin exocytosis is associated with partial restored actin dynamics and fusion events during the two phases of the secretion, with a preferential involvement of Epac2 signaling in the first phase and a rather involvement of PKA signaling in the second phase of insulin exocytosis. All these data provide some new insights into the mechanism by which current therapeutics may be improving insulin secretion.


Subject(s)
Actins/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Signal Transduction/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Cell Line, Tumor , Exocytosis/drug effects , Glucose/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Insulin-Secreting Cells/pathology , Male , Rats , Rats, Wistar
12.
J Diabetes Res ; 2013: 763125, 2013.
Article in English | MEDLINE | ID: mdl-23762875

ABSTRACT

The aim of the present study was to evaluate the potential antidiabetic effects of two-component drug Subetta and its components (release-active dilutions of antibodies to ß -subunit insulin receptor (RAD of Abs to ß -InsR) and to endothelial nitric oxide synthase (RAD of Abs to eNOS)) in Goto-Kakizaki (Paris colony) (GK/Par) diabetic rats. Subetta was administered orally for 28 days once daily (5 mL/kg) and compared to its two components (2.5 mL/kg), Rosiglitazone (5 mg/kg), and vehicle (5 mL water/kg). At day 28, fasting plasma glucose levels were significantly decreased only in Subetta and Rosiglitazone groups as compared to vehicle (P < 0.01): 147 ± 4 mg/dL and 145 ± 4 mg/dL and 165 ± 4 mg/dL, respectively. The data of glucose tolerance test showed that Subetta and RAD of Abs to ß -InsR (similar to Rosiglitazone) prevented significantly (P < 0.01) the age-related spontaneous deterioration of glucose tolerance as seen in the control group. Subetta and RAD of Abs to ß -InsR did not significantly modify the glucose-induced insulin secretion. Chronic administration of Subetta and RAD of Abs to ß -InsR improves glucose control, to an extent similar to that of Rosiglitazone. We hypothesize that Subetta and RAD of Abs to ß -InsR mostly act via an insulin-sensitizing effect upon target tissues.


Subject(s)
Antibodies/therapeutic use , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/blood , Animals , Antibodies/pharmacology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Male , Nitric Oxide Synthase Type III/antagonists & inhibitors , Rats , Receptor, Insulin/antagonists & inhibitors , Rosiglitazone , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
13.
Mol Ther ; 20(10): 1944-52, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22828498

ABSTRACT

Endocrine and exocrine insufficiencies are associated with serious diseases such as diabetes and pancreatitis, respectively. Pancreatic cells retain the capacity to regenerate in the context of cell deficiency. The remnant pancreas after pancreatectomy (Px) is a valuable target for testing the efficiency of pharmacological interventions to stimulate cell regeneration. Here, we tested the ability of GSK3ß downregulation on the stimulation of ß- and acinar cell regeneration after 90% Px in adult rats. We developed an in vivo approach based on local silencing of GSK3ß, by delivering antisense morpholino-oligonucleotides within the remnant pancreas of 90% pancreatectomized rats, and evaluated its impact on the regenerative potential of pancreatic ß and exocrine cells. ß-Cell (BC) mass was evaluated by morphometry. Cell proliferation and apoptosis were assessed by 5'bromo 2'deoxyuridine (BrdU) incorporation method and TUNEL assay, respectively. The expression of Sox9, Neurogenin-3 (Ngn3), and PDX1 was evaluated by immunohistochemistry. We show that intrapancreatic GSK3ß knockdown leads to increased BC mass (BCM) in 90% pancreatectomized rats by promoting both BC proliferation and differentiation. Moreover, downregulation of GSK3ß significantly improves exocrine growth and prevents acinar cell apoptosis in vivo. Our study designates GSK3ß as a viable drug target for therapeutic intervention on diseases of endocrine and exocrine pancreas associated with cell deficiency.


Subject(s)
Acinar Cells/physiology , Glycogen Synthase Kinase 3/genetics , Insulin-Secreting Cells/physiology , Pancreas/physiology , Regeneration , Acinar Cells/cytology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine/analysis , Cell Differentiation , Cell Proliferation , Down-Regulation , Gene Knockdown Techniques/methods , Glycogen Synthase Kinase 3 beta , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Nick-End Labeling/methods , Insulin-Secreting Cells/cytology , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas, Exocrine/cytology , Pancreas, Exocrine/physiology , Pancreatectomy/methods , Rats , Rats, Wistar , Trans-Activators/genetics , Trans-Activators/metabolism
14.
Antioxid Redox Signal ; 17(3): 433-44, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22229526

ABSTRACT

AIMS: Hypothalamic mitochondrial reactive oxygen species (mROS)-mediated signaling has been recently shown to be involved in the regulation of energy homeostasis. However, the upstream signals that control this mechanism have not yet been determined. Here, we hypothesize that glucose-induced mitochondrial fission plays a significant role in mROS-dependent hypothalamic glucose sensing. RESULTS: Glucose-triggered translocation of the fission protein dynamin-related protein 1 (DRP1) to mitochondria was first investigated in vivo in hypothalamus. Thus, we show that intracarotid glucose injection induces the recruitment of DRP1 to VMH mitochondria in vivo. Then, expression was transiently knocked down by intra-ventromedial hypothalamus (VMH) DRP1 siRNA (siDRP1) injection. 72 h post siRNA injection, brain intracarotid glucose induced insulin secretion, and VMH glucose infusion-induced refeeding decrease were measured, as well as mROS production. The SiDRP1 rats decreased mROS and impaired intracarotid glucose injection-induced insulin secretion. In addition, the VMH glucose infusion-induced refeeding decrease was lost in siDRP1 rats. Finally, mitochondrial function was evaluated by oxygen consumption measurements after DRP1 knock down. Although hypothalamic mitochondrial respiration was not modified in the resting state, substrate-driven respiration was impaired in siDRP1 rats and associated with an alteration of the coupling mechanism. INNOVATION AND CONCLUSION: Collectively, our results suggest that glucose-induced DRP1-dependent mitochondrial fission is an upstream regulator for mROS signaling, and consequently, a key mechanism in hypothalamic glucose sensing. Thus, for the first time, we demonstrate the involvement of DRP1 in physiological regulation of brain glucose-induced insulin secretion and food intake inhibition. Such involvement implies DRP1-dependent mROS production.


Subject(s)
Arcuate Nucleus of Hypothalamus/enzymology , Dynamins/metabolism , Glucose/metabolism , Mitochondria/enzymology , Ventromedial Hypothalamic Nucleus/enzymology , Animals , Appetite Regulation , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/ultrastructure , Dynamins/genetics , Energy-Generating Resources , Gene Knockdown Techniques , Glucose/physiology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Male , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/enzymology , Oxygen Consumption , Protein Transport , RNA Interference , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/ultrastructure
15.
Mol Cell Endocrinol ; 350(1): 136-44, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22209745

ABSTRACT

Serotonin and insulin are key regulators of homeostatic mechanisms in the hypothalamus. However, in type 2 diabetes, the hypothalamic responsiveness to serotonin is not clearly established. We used a diabetic model, the Goto Kakizaki (GK) rats, to explore insulin receptor expression, insulin and serotonin efficiency in the hypothalamus and liver by means of Akt phosphorylation. Insulin or dexfenfluramine (stimulator of serotonin) treatment induced Akt phosphorylation in Wistar rats but not in GK rats that exhibit down-regulated insulin receptor. Studies in a neuroblastoma cell line showed that serotonin-induced Akt phosphorylation is PI3-kinase dependent. Finally, in response to food intake, hypothalamic serotonin release was reduced in GK rats, indicating impaired responsiveness of this neurotransmitter. In conclusion, hypothalamic serotonin as insulin efficiency is impaired in diabetic GK rats. The insulin-serotonin cross-talk and impairment observed is one potential key modification in the brain during the onset of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/blood , Insulin/blood , Receptor Cross-Talk , Serotonin/blood , Animals , Blood Glucose , Cell Line, Tumor , Dexfenfluramine/pharmacology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Eating , Humans , Hypothalamus/enzymology , Hypothalamus/metabolism , Insulin/pharmacology , Insulin/physiology , Leptin/blood , Liver/enzymology , Liver/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Postprandial Period , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptor, Insulin/metabolism , Serotonin/pharmacology , Serotonin/physiology , Serotonin Receptor Agonists/pharmacology
16.
Am J Physiol Endocrinol Metab ; 301(5): E797-806, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21750265

ABSTRACT

cAMP-raising agents with glucagon-like peptide-1 (GLP-1) as the first in class, exhibit multiple actions that are beneficial for the treatment of type 2 diabetic (T2D) patients, including improvement of glucose-induced insulin secretion (GIIS). To gain additional insight into the role of cAMP in the disturbed stimulus-secretion coupling within the diabetic ß-cell, we examined more thoroughly the relationship between changes in islet cAMP concentration and insulin release in the GK/Par rat model of T2D. Basal cAMP content in GK/Par islets was significantly higher, whereas their basal insulin release was not significantly different from that of Wistar (W) islets. Even in the presence of IBMX or GLP-1, their insulin release did not significantly change despite further enhanced cAMP accumulation in both cases. The high basal cAMP level most likely reflects an increased cAMP generation in GK/Par compared with W islets since 1) forskolin dose-dependently induced an exaggerated cAMP accumulation; 2) adenylyl cyclase (AC)2, AC3, and G(s)α proteins were overexpressed; 3) IBMX-activated cAMP accumulation was less efficient and PDE-3B and PDE-1C mRNA were decreased. Moreover, the GK/Par insulin release apparatus appears less sensitive to cAMP, since GK/Par islets released less insulin at submaximal cAMP levels and required five times more cAMP to reach a maximal secretion rate no longer different from W. GLP-1 was able to reactivate GK/Par insulin secretion so that GIIS became indistinguishable from that of W. The exaggerated cAMP production is instrumental, since GLP-1-induced GIIS reactivation was lost in the presence the AC blocker 2',5'-dideoxyadenosine. This GLP-1 effect takes place in the absence of any improvement of the [Ca(2+)](i) response and correlates with activation of the cAMP-dependent PKA-dependent pathway.


Subject(s)
Cyclic AMP/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Male , Rats , Rats, Wistar , Secretory Pathway/drug effects , Secretory Pathway/physiology , Streptozocin
17.
Biochem J ; 438(1): 177-89, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21592087

ABSTRACT

Pancreatic ß-cell apoptosis induced by palmitate requires high glucose concentrations. Ceramides have been suggested to be important mediators of glucolipotoxicity-induced ß-cell apoptosis. In INS-1 ß-cells, 0.4 mM palmitate with 5 mM glucose increased the levels of dihydrosphingosine and dihydroceramides, two lipid intermediates in the de novo biosynthesis of ceramides, without inducing apoptosis. Increasing glucose concentrations to 30 mM amplified palmitate-induced accumulation of dihydrosphingosine and the formation of (dihydro)ceramides. Of note, glucolipotoxicity specifically induced the formation of C(18:0), C(22:0) and C(24:1) (dihydro)ceramide molecular species, which was associated with the up-regulation of CerS4 (ceramide synthase 4) levels. Fumonisin-B1, a ceramide synthase inhibitor, partially blocked apoptosis induced by glucolipotoxicity. In contrast, apoptosis was potentiated in the presence of D,L-threo-1-phenyl-2-palmitoylamino-3-morpholinopropan-1-ol, an inhibitor of glucosylceramide synthase. Moreover, overexpression of CerS4 amplified ceramide production and apoptosis induced by palmitate with 30 mM glucose, whereas down-regulation of CerS4 by siRNA (short interfering RNA) reduced apoptosis. CerS4 also potentiates ceramide accumulation and apoptosis induced by another saturated fatty acid: stearate. Collectively, our results suggest that glucolipotoxicity induces ß-cell apoptosis through a dual mechanism involving de novo ceramide biosynthesis and the formation of ceramides with specific N-acyl chain lengths rather than an overall increase in ceramide content.


Subject(s)
Apoptosis/drug effects , Ceramides/metabolism , Fumonisins/toxicity , Glucose/toxicity , Insulin-Secreting Cells/metabolism , Oxidoreductases/metabolism , Palmitates/toxicity , Animals , Blotting, Western , Cell Proliferation/drug effects , Enzyme Inhibitors/toxicity , Humans , Insulin-Secreting Cells/drug effects , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sweetening Agents/toxicity
18.
PLoS One ; 4(8): e6500, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19654863

ABSTRACT

BACKGROUND: Oxidative stress (OS), through excessive and/or chronic reactive oxygen species (ROS), is a mediator of diabetes-related damages in various tissues including pancreatic beta-cells. Here, we have evaluated islet OS status and beta-cell response to ROS using the GK/Par rat as a model of type 2 diabetes. METHODOLOGY/PRINCIPAL FINDINGS: Localization of OS markers was performed on whole pancreases. Using islets isolated from 7-day-old or 2.5-month-old male GK/Par and Wistar control rats, 1) gene expression was analyzed by qRT-PCR; 2) insulin secretion rate was measured; 3) ROS accumulation and mitochondrial polarization were assessed by fluorescence methods; 4) antioxidant contents were quantified by HPLC. After diabetes onset, OS markers targeted mostly peri-islet vascular and inflammatory areas, and not islet cells. GK/Par islets revealed in fact protected against OS, because they maintained basal ROS accumulation similar or even lower than Wistar islets. Remarkably, GK/Par insulin secretion also exhibited strong resistance to the toxic effect of exogenous H(2)O(2) or endogenous ROS exposure. Such adaptation was associated to both high glutathione content and overexpression (mRNA and/or protein levels) of a large set of genes encoding antioxidant proteins as well as UCP2. Finally, we showed that such a phenotype was not innate but spontaneously acquired after diabetes onset, as the result of an adaptive response to the diabetic environment. CONCLUSIONS: The GK/Par model illustrates the effectiveness of adaptive response to OS by beta-cells to achieve self-tolerance. It remains to be determined to what extend such islet antioxidant defenses upregulation might contribute to GK/Par beta-cell secretory dysfunction.


Subject(s)
Adaptation, Physiological , Diabetes Mellitus, Experimental/pathology , Islets of Langerhans/physiopathology , Oxidative Stress , Up-Regulation , Animals , Diabetes Mellitus, Experimental/metabolism , Hydrogen Peroxide/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Male , Rats , Rats, Wistar
19.
Am J Physiol Lung Cell Mol Physiol ; 295(3): L489-96, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18586953

ABSTRACT

Particulate atmospheric pollutants interact with the human airway epithelium, which releases cytokines, chemokines, and EGF receptor (EGFR) ligands leading to proinflammatory responses. There is little information concerning the short-term effects of EGFR activation by extracellular ligands on ionic regulation of airway surface lining fluids. We identified in the membrane of human epithelial bronchial cells (16HBE14o(-) line) an endogenous calcium- and voltage-dependent, outwardly rectifying small-conductance chloride channel (CACC), and we examined the effects of EGF on CACC activity. Ion channel currents were recorded with the patch-clamp technique. In cell-attached membrane patches, CACC were activated by exposure of the external surface of the cells to physiological concentrations of EGF without any change in cytosolic Ca(2+) concentration ([Ca(2+)](i)) and inhibited by tyrphostin AG-1478 (an inhibitor of EGFR that also blocks EGF-dependent Src family kinase activation). EGF activation of c-Src protein in 16HBE14o(-) cells was observed, and the signaling pathway elicited by EGFR was blocked by tyrphostin AG-1478. In excised inside-out membrane patches CACC were activated by exposure of the cytoplasmic face of the channels to the human recombinant Src(p60(c-src)) kinase with endogenous or exogenous ATP and inhibited by lambda-protein phosphatase. Secretion of EGFR ligands by epithelial airway cells exposed to pollutants would then elicit a rapid and direct ionic response of CACC mediated by EGFR activation via a Src kinase family-dependent signaling pathway.


Subject(s)
Bronchi/metabolism , Chloride Channels/metabolism , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Adenosine Triphosphate/metabolism , Bronchi/cytology , Bronchi/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Humans , Patch-Clamp Techniques , Proto-Oncogene Proteins pp60(c-src)/genetics , Quinazolines , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology
20.
Diabetes ; 54(11): 3229-37, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16249449

ABSTRACT

Because acetylcholine (ACh) is a recognized potentiator of glucose-stimulated insulin release in the normal beta-cell, we have studied ACh's effect on islets of the Goto-Kakizaki (GK) rat, a spontaneous model of type 2 diabetes. We first verified that ACh was able to restore the insulin secretory glucose competence of the GK beta-cell. Then, we demonstrated that in GK islets 1) ACh elicited a first-phase insulin release at low glucose, whereas it had no effect in Wistar; 2) total phospholipase C activity, ACh-induced inositol phosphate production, and intracellular free calcium concentration ([Ca2+]i) elevation were normal; 3) ACh triggered insulin release, even in the presence of thapsigargin, which induced a reduction of the ACh-induced [Ca2+]i response (suggesting that ACh produces amplification signals that augment the efficacy of elevated [Ca2+]i on GK exocytosis); 4) inhibition of protein kinase C did not affect [Ca2+]i nor the insulin release responses to ACh; and 5) inhibition of cAMP-dependent protein kinases (PKAs), adenylyl cyclases, or cAMP generation, while not affecting the [Ca2+]i response, significantly lowered the insulinotropic response to ACh (at low and high glucose). In conclusion, ACh acts mainly through activation of the cAMP/PKA pathway to potently enhance Ca2+-stimulated insulin release in the GK beta-cell and, in doing so, normalizes its defective glucose responsiveness.


Subject(s)
Acetylcholine/pharmacology , Cyclic AMP/biosynthesis , Glucose/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Muscarinic/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Insulin Secretion , Male , Rats , Signal Transduction/drug effects , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL