Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Endocrinol Metab ; 317(2): E234-E243, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31013146

RESUMO

The pancreatic ß-cell responds to changes in the nutrient environment to maintain glucose homeostasis by adapting its function and mass. Nutrients can act directly on the ß-cell and also indirectly through the brain via autonomic nerves innervating islets. Despite the importance of the brain-islet axis in insulin secretion, relatively little is known regarding its involvement in ß-cell proliferation. We previously demonstrated that prolonged infusions of nutrients in rats provoke a dramatic increase in ß-cell proliferation in part because of the direct action of nutrients. Here, we addressed the contribution of the autonomic nervous system. In isolated islets, muscarinic stimulation increased, whereas adrenergic stimulation decreased, glucose-induced ß-cell proliferation. Blocking α-adrenergic receptors reversed the effect of epinephrine on glucose + nonesterified fatty acids (NEFA)-induced ß-cell proliferation, whereas activation of ß-adrenergic receptors was without effect. Infusion of glucose + NEFA toward the brain stimulated ß-cell proliferation, and this effect was abrogated following celiac vagotomy. The increase in ß-cell proliferation following peripheral infusions of glucose + NEFA was not inhibited by vagotomy or atropine treatment but was blocked by coinfusion of epinephrine. We conclude that ß-cell proliferation is stimulated by parasympathetic and inhibited by sympathetic signals. Whereas glucose + NEFA in the brain stimulates ß-cell proliferation through the vagus nerve, ß-cell proliferation in response to systemic nutrient excess does not involve parasympathetic signals but may be associated with decreased sympathetic tone.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Proliferação de Células , Células Secretoras de Insulina/fisiologia , Envelhecimento/fisiologia , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Glicemia/metabolismo , Carbacol/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Epinefrina/farmacologia , Ácidos Graxos não Esterificados/farmacologia , Glucose/farmacologia , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos Lew
2.
Diabetologia ; 61(3): 526-538, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29143855

RESUMO

Mice are the most commonly used species in preclinical research on the pathophysiology of metabolic diseases. Although they are extremely useful for identifying pathways, mechanisms and genes regulating glucose and energy homeostasis, the specificities of the various mouse models and methodologies used to investigate a metabolic phenotype can have a profound impact on experimental results and their interpretation. This review aims to: (1) describe the most commonly used experimental tests to assess glucose and energy homeostasis in mice; (2) provide some guidelines regarding the design, analysis and interpretation of these tests, as well as for studies using genetic models; and (3) identify important caveats and confounding factors that must be taken into account in the interpretation of findings.


Assuntos
Diabetes Mellitus/metabolismo , Animais , Diabetes Mellitus/patologia , Modelos Animais de Doenças , Metabolismo Energético/fisiologia , Glucose/metabolismo , Camundongos
3.
Am J Physiol Endocrinol Metab ; 315(6): E1251-E1263, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30106624

RESUMO

Pancreatic ß-cell expansion is a highly regulated metabolic adaptation to increased somatic demands, including obesity and pregnancy; adult ß cells otherwise rarely proliferate. We previously showed that high-fat diet (HFD) feeding induces mouse ß-cell proliferation in less than 1 wk in the absence of insulin resistance. Here we metabolically profiled tissues from a short-term HFD ß-cell expansion mouse model to identify pathways and metabolite changes associated with ß-cell proliferation. Mice fed HFD vs. chow diet (CD) showed a 14.3% increase in body weight after 7 days; ß-cell proliferation increased 1.75-fold without insulin resistance. Plasma from 1-wk HFD-fed mice induced ß-cell proliferation ex vivo. The plasma, as well as liver, skeletal muscle, and bone, were assessed by LC and GC mass-spectrometry for global metabolite changes. Of the 1,283 metabolites detected, 159 showed significant changes [false discovery rate (FDR) < 0.1]. The majority of changes were in liver and muscle. Pathway enrichment analysis revealed key metabolic changes in steroid synthesis and lipid metabolism, including free fatty acids and other bioactive lipids. Other important enrichments included changes in the citric acid cycle and 1-carbon metabolism pathways implicated in DNA methylation. Although the minority of changes were observed in bone and plasma (<20), increased p-cresol sulfate was increased >4 fold in plasma (the largest increase in all tissues), and pantothenate (vitamin B5) decreased >2-fold. The results suggest that HFD-mediated ß-cell expansion is associated with complex, global metabolite changes. The finding could be a significant insight into Type 2 diabetes pathogenesis and potential novel drug targets.


Assuntos
Proliferação de Células/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica , Células Secretoras de Insulina/citologia , Lipídeos/sangue , Animais , Glicemia , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Obesidade/metabolismo
4.
Diabetologia ; 60(5): 879-888, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28078385

RESUMO

AIMS/HYPOTHESIS: The mechanisms underlying pancreatic islet mass expansion have attracted considerable interest as potential therapeutic targets to prevent or delay the onset of type 2 diabetes. While several factors promoting beta cell proliferation have been identified, in the context of nutrient excess the roles of glucose or NEFA in relation to insulin resistance remain unclear. Here we tested the hypothesis that glucose and NEFA synergistically and reversibly promote beta cell proliferation in the context of nutrient-induced insulin resistance. METHODS: Using 72 h infusions of glucose (GLU) or the oleate-enriched lipid emulsion ClinOleic (CLI), singly or in combination, we assessed beta cell proliferation, islet mass and insulin sensitivity in male Lewis rats. The effects of nutrients and endogenous circulating factors were examined in isolated and transplanted islets. Reversibility was studied 3 and 6 days after the end of the infusion. RESULTS: GLU infusions modestly stimulated beta cell proliferation, CLI alone had no effect and GLU+CLI infusions markedly stimulated beta cell proliferation. Insulin sensitivity was equally decreased in GLU and GLU+CLI infusions. GLU+CLI infusions also stimulated beta cell proliferation in islets transplanted under the kidney capsule, albeit to a lesser extent compared with endogenous islets. Ex vivo, the combination of glucose and NEFA enhanced beta cell proliferation in rat and human islets independently from secreted insulin, and serum from GLU+CLI-infused rats potentiated the effect of glucose. Glucose tolerance, beta cell proliferation and islet mass were all restored to normal levels 6 days after termination of the infusion. CONCLUSIONS/INTERPRETATION: Glucose and NEFA synergistically and reversibly promote beta cell proliferation in part via direct action on the beta cell and independently from secreted insulin.


Assuntos
Ácidos Graxos/farmacologia , Glucose/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Ácidos Graxos/administração & dosagem , Glucose/administração & dosagem , Técnica Clamp de Glucose , Técnicas In Vitro , Masculino , Distribuição Aleatória , Ratos
5.
Handb Exp Pharmacol ; 236: 159-180, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27807697

RESUMO

Of the 415 million people suffering from diabetes worldwide, 90% have type 2 diabetes. Type 2 diabetes is characterized by hyperglycemia and occurs in obese individuals as a result of insulin resistance and inadequate insulin levels. Accordingly, diabetes drugs are tailored to enhance glucose disposal or target the pancreatic islet ß cell to increase insulin secretion. The majority of the present-day insulin secretagogues, however, increase the risk of iatrogenic hypoglycemia, and hence alternatives are actively sought. The long-chain fatty acid, G protein-coupled receptor FFA1/Gpr40, is expressed in ß cells, and its activation potentiates insulin secretion in a glucose-dependent manner. Preclinical data indicate that FFA1 agonism is an effective treatment to restore glucose homeostasis in rodent models of diabetes. This initial success prompted clinical trials in type 2 diabetes patients, the results of which were promising; however, the field suffered a significant setback when the lead compound TAK-875/fasiglifam was withdrawn from clinical development due to liver safety concerns. Nevertheless, recent developments have brought to light a surprising complexity of FFA1 agonist action, signaling diversity, and biological outcomes, raising hopes that with a greater understanding of the mechanisms at play the second round will be more successful.


Assuntos
Receptores Acoplados a Proteínas G/fisiologia , Animais , Benzofuranos/efeitos adversos , Benzofuranos/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/fisiologia , Doenças Metabólicas/etiologia , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais , Sulfonas/efeitos adversos , Sulfonas/uso terapêutico
6.
J Biol Chem ; 290(34): 21131-21140, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26157145

RESUMO

FFAR1/GPR40 is a seven-transmembrane domain receptor (7TMR) expressed in pancreatic ß cells and activated by FFAs. Pharmacological activation of GPR40 is a strategy under consideration to increase insulin secretion in type 2 diabetes. GPR40 is known to signal predominantly via the heterotrimeric G proteins Gq/11. However, 7TMRs can also activate functionally distinct G protein-independent signaling via ß-arrestins. Further, G protein- and ß-arrestin-based signaling can be differentially modulated by different ligands, thus eliciting ligand-specific responses ("biased agonism"). Whether GPR40 engages ß-arrestin-dependent mechanisms and is subject to biased agonism is unknown. Using bioluminescence resonance energy transfer-based biosensors for real-time monitoring of cell signaling in living cells, we detected a ligand-induced GPR40-ß-arrestin interaction, with the synthetic GPR40 agonist TAK-875 being more effective than palmitate or oleate in recruiting ß-arrestins 1 and 2. Conversely, TAK-875 acted as a partial agonist of Gq/11-dependent GPR40 signaling relative to both FFAs. Pharmacological blockade of Gq activity decreased FFA-induced insulin secretion. In contrast, knockdown or genetic ablation of ß-arrestin 2 in an insulin-secreting cell line and mouse pancreatic islets, respectively, uniquely attenuated the insulinotropic activity of TAK-875, thus providing functional validation of the biosensor data. Collectively, these data reveal that in addition to coupling to Gq/11, GPR40 is functionally linked to a ß-arrestin 2-mediated insulinotropic signaling axis. These observations expose previously unrecognized complexity for GPR40 signal transduction and may guide the development of biased agonists showing improved clinical profile in type 2 diabetes.


Assuntos
Arrestinas/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Animais , Arrestinas/antagonistas & inibidores , Arrestinas/metabolismo , Benzofuranos/farmacologia , Técnicas Biossensoriais , Linhagem Celular Tumoral , Espectroscopia de Ressonância de Spin Eletrônica , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Insulina/agonistas , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Cinética , Camundongos , Ácido Oleico/farmacologia , Ácido Palmítico/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Sulfonas/farmacologia , Técnicas de Cultura de Tecidos , beta-Arrestina 2 , beta-Arrestinas
7.
Artigo em Inglês | MEDLINE | ID: mdl-26888796

RESUMO

BACKGROUND: GPR120 (FFAR4) is a G-protein coupled receptor implicated in the development of obesity and the antiinflammatory and insulin-sensitizing effects of omega-3 (ω-3) polyunsaturated fatty acids. Increasing central ω-3 polyunsaturated fatty acid levels has been shown to have both anorectic and anxiolytic actions. Despite the strong clinical interest in GPR120, its role in the brain is largely unknown, and thus we sought to determine the impact of central GPR120 pharmacological activation on energy balance, food reward, and anxiety-like behavior. METHODS: Male C57Bl/6 mice with intracerebroventricular cannulae received a single injection (0.1 or 1 µM) or continuous 2-week infusion (1 µM/d; mini-pump) of a GPR120 agonist or vehicle. Free-feeding intake, operant lever-pressing for palatable food, energy expenditure (indirect calorimetry), and body weight were measured. GPR120 mRNA expression was measured in pertinent brain areas. Anxiety-like behavior was assessed in the elevated-plus maze and open field test. RESULTS: GPR120 agonist injections substantially reduced chow intake during 4 hours postinjection, suppressed the rewarding effects of high-fat/-sugar food, and blunted approach-avoidance behavior in the open field. Conversely, prolonged central GPR120 agonist infusions reduced anxiety-like behavior in the elevated-plus maze and open field, yet failed to affect free-feeding intake, energy expenditure, and body weight on a high-fat diet. CONCLUSION: Acute reductions in food intake and food reward suggest that GPR120 could mediate the effects of central ω-3 polyunsaturated fatty acids to inhibit appetite. The anxiolytic effect elicited by GPR120 agonist infusions favors the testing of compounds that can enter the brain to activate GPR120 for the mitigation of anxiety.


Assuntos
Ansiedade/prevenção & controle , Ingestão de Alimentos/fisiologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiologia , Recompensa , Animais , Benzofuranos/administração & dosagem , Benzofuranos/farmacologia , Peso Corporal/efeitos dos fármacos , Condicionamento Operante/fisiologia , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético/efeitos dos fármacos , Infusões Intraventriculares , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Atividade Motora/efeitos dos fármacos , Receptores Acoplados a Proteínas G/biossíntese , Sulfonas/administração & dosagem , Sulfonas/farmacologia
8.
Am J Physiol Endocrinol Metab ; 308(7): E573-82, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25628421

RESUMO

Both short- (1 wk) and long-term (2-12 mo) high-fat diet (HFD) studies reveal enhanced ß-cell mass due to increased ß-cell proliferation. ß-Cell proliferation following HFD has been postulated to occur in response to insulin resistance; however, whether HFD can induce ß-cell proliferation independent of insulin resistance has been controversial. To examine the kinetics of HFD-induced ß-cell proliferation and its correlation with insulin resistance, we placed 8-wk-old male C57Bl/6J mice on HFD for different lengths of time and assayed the following: glucose tolerance, insulin secretion in response to glucose, insulin tolerance, ß-cell mass, and ß-cell proliferation. We found that ß-cell proliferation was significantly increased after only 3 days of HFD feeding, weeks before an increase in ß-cell mass or peripheral insulin resistance was detected. These results were confirmed by hyperinsulinemic euglycemic clamps and measurements of α-hydroxybutyrate, a plasma biomarker of insulin resistance in humans. An increase in expression of key islet-proliferative genes was found in isolated islets from 1-wk HFD-fed mice compared with chow diet (CD)-fed mice. These data indicate that short-term HFD feeding enhances ß-cell proliferation before insulin resistance becomes apparent.


Assuntos
Proliferação de Células , Dieta Hiperlipídica , Resistência à Insulina , Células Secretoras de Insulina/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/farmacologia , Técnica Clamp de Glucose , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Teste de Tolerância a Glucose , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 109(7): 2376-81, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308370

RESUMO

The G protein-coupled free fatty acid receptor-1 (FFA1/GPR40) plays a major role in the regulation of insulin secretion by fatty acids. GPR40 is considered a potential therapeutic target to enhance insulin secretion in type 2 diabetes; however, its mode of regulation is essentially unknown. The aims of this study were to test the hypothesis that glucose regulates GPR40 gene expression in pancreatic ß-cells and to determine the mechanisms of this regulation. We observed that glucose stimulates GPR40 gene transcription in pancreatic ß-cells via increased binding of pancreas-duodenum homeobox-1 (Pdx-1) to the A-box in the HR2 region of the GPR40 promoter. Mutation of the Pdx-1 binding site within the HR2 abolishes glucose activation of GPR40 promoter activity. The stimulation of GPR40 expression and Pdx-1 binding to the HR2 in response to glucose are mimicked by N-acetyl glucosamine, an intermediate of the hexosamine biosynthesis pathway, and involve PI3K-dependent O-GlcNAcylation of Pdx-1 in the nucleus. We demonstrate that O-GlcNAc transferase (OGT) interacts with the product of the PI3K reaction, phosphatidylinositol 3,4,5-trisphosphate (PIP(3)), in the nucleus. This interaction enables OGT to catalyze O-GlcNAcylation of nuclear proteins, including Pdx-1. We conclude that glucose stimulates GPR40 gene expression at the transcriptional level through Pdx-1 binding to the HR2 region and via a signaling cascade that involves an interaction between OGT and PIP(3) at the nuclear membrane. These observations reveal a unique mechanism by which glucose metabolism regulates the function of transcription factors in the nucleus to induce gene expression.


Assuntos
Acetilglucosamina/metabolismo , Duodeno/metabolismo , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Pâncreas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Acoplados a Proteínas G/genética , Transcrição Gênica , Animais , Hexosaminas/biossíntese , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Camundongos Endogâmicos C57BL
11.
J Biol Chem ; 288(34): 24825-33, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23853095

RESUMO

In pancreatic ß-cells, glucose induces the binding of the transcription factor pancreatic duodenal homeobox-1 (PDX-1) to the insulin gene promoter to activate insulin gene transcription. At low glucose levels, glycogen synthase kinase 3ß (GSK3ß) is known to phosphorylate PDX-1 on C-terminal serine residues, which triggers PDX-1 proteasomal degradation. We previously showed that the serine/threonine Per-Arnt-Sim domain-containing kinase (PASK) regulates insulin gene transcription via PDX-1. However, the mechanisms underlying this regulation are unknown. In this study, we aimed to identify the role of PASK in the regulation of PDX-1 phosphorylation, protein expression, and stability in insulin-secreting cells and isolated rodent islets of Langerhans. We observed that glucose induces a decrease in overall PDX-1 serine phosphorylation and that overexpression of WT PASK mimics this effect. In vitro, PASK directly phosphorylates GSK3ß on its inactivating phosphorylation site Ser(9). Overexpression of a kinase-dead (KD), dominant negative version of PASK blocks glucose-induced Ser(9) phosphorylation of GSK3ß. Accordingly, GSK3ß Ser(9) phosphorylation is reduced in islets from pask-null mice. Overexpression of WT PASK or KD GSK3ß protects PDX-1 from degradation and results in increased PDX-1 protein abundance. Conversely, overexpression of KD PASK blocks glucose-induction of PDX-1 protein. We conclude that PASK phosphorylates and inactivates GSK3ß, thereby preventing PDX-1 serine phosphorylation and alleviating GSK3ß-mediated PDX-1 protein degradation in pancreatic ß-cells.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Animais , Glucose/farmacologia , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Células Hep G2 , Proteínas de Homeodomínio/genética , Humanos , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Mutação , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ratos Wistar , Edulcorantes/farmacologia , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
12.
J Biol Chem ; 288(9): 6542-51, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23335512

RESUMO

The mechanisms linking fat intake to bone loss remain unclear. By demonstrating the expression of the free fatty acid receptor G-coupled protein receptor 40 (GPR40) in bone cells, we hypothesized that this receptor may play a role in mediating the effects of fatty acids on bone remodeling. Using micro-CT analysis, we showed that GPR40(-/-) mice exhibit osteoporotic features suggesting a positive role of GPR40 on bone density. In primary cultures of bone marrow, we showed that GW9508, a GRP40 agonist, abolished bone-resorbing cell differentiation. This alteration of the receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation occurred via the inhibition of the nuclear factor κB (NF-κB) signaling pathway as demonstrated by decrease in gene reporter activity, inhibitor of κB kinase (IKKα/ß) activation, inhibitor of κB (IkBα) phosphorylation, and nuclear factor of activated T cells 1 (NFATc1) expression. The GPR40-dependent effect of GW9508 was confirmed using shRNA interference in osteoclast precursors and GPR40(-/-) primary cell cultures. In addition, in vivo administration of GW9508 counteracted ovariectomy-induced bone loss in wild-type but not GPR40(-/-) mice, enlightening the obligatory role of the GPR40 receptor. Then, in a context of growing prevalence of metabolic and age-related bone disorders, our results demonstrate for the first time in translational approaches that GPR40 is a relevant target for the design of new nutritional and therapeutic strategies to counter bone complications.


Assuntos
Reabsorção Óssea/metabolismo , Diferenciação Celular , Osteoclastos/metabolismo , Osteoporose/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Reabsorção Óssea/dietoterapia , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Linhagem Celular , Metilaminas/farmacologia , Camundongos , Camundongos Knockout , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/patologia , Osteoporose/dietoterapia , Osteoporose/genética , Osteoporose/patologia , Propionatos/farmacologia , Ligante RANK/genética , Ligante RANK/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
13.
Arterioscler Thromb Vasc Biol ; 33(5): 954-61, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23520164

RESUMO

OBJECTIVE: Nitro-oxidative stress exerts a significant role in the genesis of hypoxic-ischemic (HI) brain injury. We previously reported that the ω-6 long chain fatty acids, transarachidonic acids (TAAs), which are nitrative stress-induced nonenzymatically generated arachidonic acid derivatives, trigger selective microvascular endothelial cell death in neonatal neural tissue. The primary molecular target of TAAs remains unidentified. GPR40 is a G protein-coupled receptor activated by long chain fatty acids, including ω-6; it is highly expressed in brain, but its functions in this tissue are largely unknown. We hypothesized that TAAs play a significant role in neonatal HI-induced cerebral microvascular degeneration through GPR40 activation. APPROACH AND RESULTS: Within 24 hours of a HI insult to postnatal day 7 rat pups, a cerebral infarct and a 40% decrease in cerebrovascular density was observed. These effects were associated with an increase in nitrative stress markers (3-nitrotyrosine immunoreactivity and TAA levels) and were reduced by treatment with nitric oxide synthase inhibitor. GPR40 was expressed in rat pup brain microvasculature. In vitro, in GPR40-expressing human embryonic kidney (HEK)-293 cells, [(14)C]-14E-AA (radiolabeled TAA) bound specifically, and TAA induced calcium transients, extracellular signal-regulated kinase 1/2 phosphorylation, and proapoptotic thrombospondin-1 expression. In vivo, intracerebroventricular injection of TAAs triggered thrombospondin-1 expression and cerebral microvascular degeneration in wild-type mice, but not in GPR40-null congeners. Additionally, HI-induced neurovascular degeneration and cerebral infarct were decreased in GPR40-null mice. CONCLUSIONS: GPR40 emerges as the first identified G protein-coupled receptor conveying actions of nonenzymatically generated nitro-oxidative products, specifically TAAs, and is involved in (neonatal) HI encephalopathy.


Assuntos
Ácido Araquidônico/metabolismo , Infarto Cerebral/etiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Células Endoteliais/fisiologia , Feminino , Células HEK293 , Humanos , Hipóxia-Isquemia Encefálica/complicações , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
14.
Med Sci (Paris) ; 29(8-9): 715-21, 2013.
Artigo em Francês | MEDLINE | ID: mdl-24005625

RESUMO

Glucose homeostasis requires a tight regulation of insulin secretion from pancreatic ß-cells. Insulin release is chiefly stimulated by glucose, but also modulated by other nutrients, including long-chain fatty acids which potentiate glucose-induced insulin secretion. The discovery of G-protein coupled receptors activated by fatty acids (and other lipid derivatives) and expressed at the surface of various cell types, including ß-cells, has added a new dimension to our understanding of the control of glucose homeostasis by fatty acids. Amongst these receptors, GPR40 and GPR119 have generated great interest as potential therapeutic targets to augment insulin secretion in type 2 diabetes. In fact, the promising results of a phase 2 clinical trial with a GPR40 agonist have provided a proof of concept for this therapeutic strategy. However, our understanding of the biology and pharmacology of these receptors remains incomplete.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Ácidos Graxos/fisiologia , Células Secretoras de Insulina/química , Receptores Acoplados a Proteínas G/fisiologia , Humanos , Células Secretoras de Insulina/fisiologia , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/análise
15.
J Endocrinol ; 258(3)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37194667

RESUMO

The year 2023 marks 100 years since publication of the first report of a hyperglycemic factor in pancreatic extracts which C P Kimball and John R Murlin named glucagon (from GLUCose AGONist). Glucagon has a range of profound effects on metabolism including, but not limited to, stimulation of hepatic glucose production. Dysregulation of glucagon secretion is a key feature of both major forms of diabetes, leading to the concept that diabetes is a bihormonal disorder. Still, the work to fully understand the production and biological effects of glucagon has proceeded at a slower pace compared to that of insulin. A recent resurgence of interest in the islet alpha (α) cell, the predominant site of glucagon production, has been facilitated in part by technological innovations. This work has led to significant developments in the field, from defining how alpha cells develop and how glucagon secretion from pancreatic alpha cells is regulated to determining the role of glucagon in metabolic homeostasis and the progression of both major forms of diabetes. In addition, glucagon is considered to be a promising target for diabetes therapy, with many new potential applications arising from research in this field. This collection of reviews, led by Guest Editors James Cantley, Vincent Poitout and Rebecca Hull-Meichle, is intended to capture the field's current understanding of glucagon and alpha cell biology, as well stimulate additional interest and research on this important hormone.


Assuntos
Células Secretoras de Glucagon , Glucagon , Glucagon/metabolismo , Aniversários e Eventos Especiais , Insulina/metabolismo , Glucose/metabolismo
16.
Diabetes ; 72(1): 45-58, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36191509

RESUMO

The functional mass of insulin-secreting pancreatic ß-cells expands to maintain glucose homeostasis in the face of nutrient excess, in part via replication of existing ß-cells. Type 2 diabetes appears when these compensatory mechanisms fail. Nutrients including glucose and fatty acids are important contributors to the ß-cell compensatory response, but their underlying mechanisms of action remain poorly understood. We investigated the transcriptional mechanisms of ß-cell proliferation in response to fatty acids. Isolated rat islets were exposed to 16.7 mmol/L glucose with or without 0.5 mmol/L oleate (C18:1) or palmitate (C16:0) for 48 h. The islet transcriptome was assessed by single-cell RNA sequencing. ß-Cell proliferation was measured by flow cytometry. Unsupervised clustering of pooled ß-cells identified different subclusters, including proliferating ß-cells. ß-Cell proliferation increased in response to oleate but not palmitate. Both fatty acids enhanced the expression of genes involved in energy metabolism and mitochondrial activity. Comparison of proliferating versus nonproliferating ß-cells and pseudotime ordering suggested the involvement of reactive oxygen species (ROS) and peroxiredoxin signaling. Accordingly, N-acetyl cysteine and the peroxiredoxin inhibitor conoidin A both blocked oleate-induced ß-cell proliferation. Our study reveals a key role for ROS signaling through peroxiredoxin activation in oleate-induced ß-cell proliferation.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Ratos , Animais , Ácidos Graxos/farmacologia , Ácidos Graxos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ácido Oleico/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Proliferação de Células , Palmitatos/metabolismo , Glucose/metabolismo , Análise de Sequência de RNA , Ilhotas Pancreáticas/metabolismo
17.
Cell Rep ; 42(5): 112500, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37171959

RESUMO

Vitamin K is a micronutrient necessary for γ-carboxylation of glutamic acids. This post-translational modification occurs in the endoplasmic reticulum (ER) and affects secreted proteins. Recent clinical studies implicate vitamin K in the pathophysiology of diabetes, but the underlying molecular mechanism remains unknown. Here, we show that mouse ß cells lacking γ-carboxylation fail to adapt their insulin secretion in the context of age-related insulin resistance or diet-induced ß cell stress. In human islets, γ-carboxylase expression positively correlates with improved insulin secretion in response to glucose. We identify endoplasmic reticulum Gla protein (ERGP) as a γ-carboxylated ER-resident Ca2+-binding protein expressed in ß cells. Mechanistically, γ-carboxylation of ERGP protects cells against Ca2+ overfilling by diminishing STIM1 and Orai1 interaction and restraining store-operated Ca2+ entry. These results reveal a critical role of vitamin K-dependent carboxylation in regulation of Ca2+ flux in ß cells and in their capacity to adapt to metabolic stress.


Assuntos
Processamento de Proteína Pós-Traducional , Vitamina K , Camundongos , Animais , Humanos , Vitamina K/farmacologia , Vitamina K/fisiologia , Osteocalcina/metabolismo , Insulina/metabolismo , Estresse Fisiológico , Cálcio/metabolismo
18.
J Proteome Res ; 11(7): 3520-32, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22578083

RESUMO

The prevalence of diabetes mellitus is increasing dramatically throughout the world, and the disease has become a major public health issue. The most common form of the disease, type 2 diabetes, is characterized by insulin resistance and insufficient insulin production from the pancreatic beta-cell. Since glucose is the most potent regulator of beta-cell function under physiological conditions, identification of the insulin secretory defect underlying type 2 diabetes requires a better understanding of glucose regulation of human beta-cell function. To this aim, a bottom-up LC-MS/MS-based proteomics approach was used to profile pooled islets from multiple donors under basal (5 mM) or high (15 mM) glucose conditions. Our analysis discovered 256 differentially abundant proteins (∼p < 0.05) after 24 h of high glucose exposure from more than 4500 identified in total. Several novel glucose-regulated proteins were elevated under high glucose conditions, including regulators of mRNA splicing (pleiotropic regulator 1), processing (retinoblastoma binding protein 6), and function (nuclear RNA export factor 1), in addition to neuron navigator 1 and plasminogen activator inhibitor 1. Proteins whose abundances markedly decreased during incubation at 15 mM glucose included Bax inhibitor 1 and synaptotagmin-17. Up-regulation of dicer 1 and SLC27A2 and down-regulation of phospholipase Cß4 were confirmed by Western blots. Many proteins found to be differentially abundant after high glucose stimulation are annotated as uncharacterized or hypothetical. These findings expand our knowledge of glucose regulation of the human islet proteome and suggest many hitherto unknown responses to glucose that require additional studies to explore novel functional roles.


Assuntos
Glucose/fisiologia , Ilhotas Pancreáticas/metabolismo , Proteoma/metabolismo , Acetiltransferases/genética , Acetiltransferases/isolamento & purificação , Acetiltransferases/metabolismo , Cromatografia por Troca Iônica , Cromatografia de Fase Reversa , Análise por Conglomerados , Coenzima A Ligases/metabolismo , RNA Helicases DEAD-box/metabolismo , Elongases de Ácidos Graxos , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/isolamento & purificação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/enzimologia , Mapeamento de Peptídeos , Fosfolipase C beta/metabolismo , Proteoma/genética , Proteoma/isolamento & purificação , Proteômica , Ribonuclease III/metabolismo , Espectrometria de Massas em Tandem , Técnicas de Cultura de Tecidos
19.
J Biol Chem ; 286(51): 44005-44014, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22065581

RESUMO

PAS kinase (PASK) is a glucose-regulated protein kinase involved in the control of pancreatic islet hormone release and insulin sensitivity. We aimed here to identify mutations in the PASK gene that may be associated with young-onset diabetes in humans. We screened 18 diabetic probands with unelucidated maturity-onset diabetes of the young (MODY). We identified two rare nonsynonymous mutations in the PASK gene (p.L1051V and p.G1117E), each of which was found in a single MODY family. Wild type or mutant PASKs were expressed in HEK 293 cells. Kinase activity of the affinity-purified proteins was assayed as autophosphorylation at amino acid Thr307 or against an Ugp1p-derived peptide. Whereas the PASK p.G1117E mutant displayed a ∼25% increase with respect to wild type PASK in the extent of autophosphorylation, and a ∼2-fold increase in kinase activity toward exogenous substrates, the activity of the p.L1051V mutant was unchanged. Amino acid Gly1117 is located in an α helical region opposing the active site of PASK and may elicit either: (a) a conformational change that increases catalytic efficiency or (b) a diminished inhibitory interaction with the PAS domain. Mouse islets were therefore infected with adenoviruses expressing wild type or mutant PASK and the regulation of insulin secretion was examined. PASK p.G1117E-infected islets displayed a 4-fold decrease in glucose-stimulated (16.7 versus 3 mM) insulin secretion, chiefly reflecting a 4.5-fold increase in insulin release at low glucose. In summary, we have characterized a rare mutation (p.G1117E) in the PASK gene from a young-onset diabetes family, which modulates glucose-stimulated insulin secretion.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Adulto , Animais , Linhagem Celular , Diabetes Mellitus/metabolismo , Genômica , Glucagon/metabolismo , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Secreção de Insulina , Masculino , Proteínas de Membrana/metabolismo , Modelos Genéticos , Mutagênese , Fosforilação , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo
20.
Endocr Rev ; 29(3): 351-66, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18048763

RESUMO

Glucotoxicity, lipotoxicity, and glucolipotoxicity are secondary phenomena that are proposed to play a role in all forms of type 2 diabetes. The underlying concept is that once the primary pathogenesis of diabetes is established, probably involving both genetic and environmental forces, hyperglycemia and very commonly hyperlipidemia ensue and thereafter exert additional damaging or toxic effects on the beta-cell. In addition to their contribution to the deterioration of beta-cell function after the onset of the disease, elevations of plasma fatty acid levels that often accompany insulin resistance may, as glucose levels begin to rise outside of the normal range, also play a pathogenic role in the early stages of the disease. Because hyperglycemia is a prerequisite for lipotoxicity to occur, the term glucolipotoxicity, rather than lipotoxicity, is more appropriate to describe deleterious effects of lipids on beta-cell function. In vitro and in vivo evidence supporting the concept of glucotoxicity is presented first, as well as a description of the underlying mechanisms with an emphasis on the role of oxidative stress. Second, we discuss the functional manifestations of glucolipotoxicity on insulin secretion, insulin gene expression, and beta-cell death, and the role of glucose in the mechanisms of glucolipotoxicity. Finally, we attempt to define the role of these phenomena in the natural history of beta-cell compensation, decompensation, and failure during the course of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/fisiologia , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Metabolismo dos Lipídeos/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa