Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
JCI Insight ; 7(21)2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36107617

RESUMO

Puberty is associated with transient insulin resistance that normally recedes at the end of puberty; however, in overweight children, insulin resistance persists, leading to an increased risk of type 2 diabetes. The mechanisms whereby pancreatic ß cells adapt to pubertal insulin resistance, and how they are affected by the metabolic status, have not been investigated. Here, we show that puberty is associated with a transient increase in ß cell proliferation in rats and humans of both sexes. In rats, ß cell proliferation correlated with a rise in growth hormone (GH) levels. Serum from pubertal rats and humans promoted ß cell proliferation, suggesting the implication of a circulating factor. In pubertal rat islets, expression of genes of the GH/serotonin (5-hydroxytryptamine [5-HT]) pathway underwent changes consistent with a proliferative effect. Inhibition of the pro-proliferative 5-HT receptor isoform HTR2B blocked the increase in ß cell proliferation in pubertal islets ex vivo and in vivo. Peripubertal metabolic stress blunted ß cell proliferation during puberty and led to altered glucose homeostasis later in life. This study identifies a role of GH/GH receptor/5-HT/HTR2B signaling in the control of ß cell mass expansion during puberty and identifies a mechanistic link between pubertal obesity and the risk of developing type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Criança , Humanos , Masculino , Feminino , Ratos , Animais , Adulto , Serotonina/metabolismo , Maturidade Sexual , Homeostase , Proliferação de Células , Glucose/metabolismo
3.
Diabetes ; 71(6): 1218-1232, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35287172

RESUMO

Fatty acid (FA) signaling contributes to ß-cell mass expansion in response to nutrient excess, but the underlying mechanisms are poorly understood. In the presence of elevated glucose, FA metabolism is shifted toward synthesis of complex lipids, including sphingolipids. Here, we tested the hypothesis that sphingolipids are involved in the ß-cell proliferative response to FA. Isolated rat islets were exposed to FA and 16.7 mmol/L glucose for 48-72 h, and the contribution of the de novo sphingolipid synthesis pathway was tested using the serine palmitoyltransferase inhibitor myriocin, the sphingosine kinase (SphK) inhibitor SKI II, or knockdown of SphK, fatty acid elongase 1 (ELOVL1) and acyl-CoA-binding protein (ACBP). Rats were infused with glucose and the lipid emulsion ClinOleic and received SKI II by gavage. ß-Cell proliferation was assessed by immunochemistry or flow cytometry. Sphingolipids were analyzed by liquid chromatography-tandem mass spectrometry. Among the FAs tested, only oleate increased ß-cell proliferation. Myriocin, SKI II, and SphK knockdown all decreased oleate-induced ß-cell proliferation. Oleate exposure did not increase the total amount of sphingolipids but led to a specific rise in 24:1 species. Knockdown of ACBP or ELOVL1 inhibited oleate-induced ß-cell proliferation. We conclude that unsaturated very-long-chain sphingolipids produced from the available C24:1 acyl-CoA pool mediate oleate-induced ß-cell proliferation in rats.


Assuntos
Ácido Oleico , Esfingolipídeos , Animais , Proliferação de Células , Ácidos Graxos/metabolismo , Glucose , Ratos , Esfingolipídeos/química
4.
Sci Rep ; 11(1): 11283, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050242

RESUMO

The potential to treat diabetes by increasing beta-cell mass is driving a major effort to identify beta-cell mitogens. Demonstration of mitogen activity in human beta cells is frequently performed in ex vivo assays. However, reported disparities in the efficacy of beta-cell mitogens led us to investigate the sources of this variability. We studied 35 male (23) and female (12) human islet batches covering a range of donor ages and BMI. Islets were kept intact or dispersed into single cells and cultured in the presence of harmine, glucose, or heparin-binding epidermal growth factor-like growth factor (HB-EGF), and subsequently analyzed by immunohistochemistry or flow cytometry. Proliferating cells were identified by double labeling with EdU and Ki67 and glucagon, c-peptide or Nkx6.1, and cytokeratin-19 to respectively label alpha, beta, and ductal cells. Harmine and HB-EGF stimulated human beta-cell proliferation, but the effect of glucose was dependent on the assay and the donor. Harmine potently stimulated alpha-cell proliferation and both harmine and HB-EGF increased proliferation of insulin- and glucagon-negative cells, including cytokeratin 19-positive cells. Given the abundance of non-beta cells in human islet preparations, our results suggest that assessment of beta-cell mitogens requires complementary approaches and rigorous identification of cell identity using multiple markers.


Assuntos
Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Mitógenos/farmacologia , Peptídeo C/metabolismo , Divisão Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Feminino , Glucagon/metabolismo , Glucose/metabolismo , Harmina/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/farmacologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Masculino , Mitógenos/imunologia , Mitógenos/metabolismo , Ductos Pancreáticos/metabolismo , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos
5.
Endocrinology ; 162(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33543237

RESUMO

The free fatty-acid receptors FFAR1 (GPR40) and FFAR4 (GPR120) are implicated in the regulation of insulin secretion and insulin sensitivity, respectively. Although GPR120 and GPR40 share similar ligands, few studies have addressed possible interactions between these 2 receptors in the control of glucose homeostasis. Here we generated mice deficient in gpr120 (Gpr120KO) or gpr40 (Gpr40KO), alone or in combination (Gpr120/40KO), and metabolically phenotyped male and female mice fed a normal chow or high-fat diet. We assessed insulin secretion in isolated mouse islets exposed to selective GPR120 and GPR40 agonists singly or in combination. Following normal chow feeding, body weight and energy intake were unaffected by deletion of either receptor, although fat mass increased in Gpr120KO females. Fasting blood glucose levels were mildly increased in Gpr120/40KO mice and in a sex-dependent manner in Gpr120KO and Gpr40KO animals. Oral glucose tolerance was slightly reduced in male Gpr120/40KO mice and in Gpr120KO females, whereas insulin secretion and insulin sensitivity were unaffected. In hyperglycemic clamps, the glucose infusion rate was lower in male Gpr120/40KO mice, but insulin and c-peptide levels were unaffected. No changes in glucose tolerance were observed in either single or double knock-out animals under high-fat feeding. In isolated islets from wild-type mice, the combination of selective GPR120 and GPR40 agonists additively increased insulin secretion. We conclude that while simultaneous activation of GPR120 and GPR40 enhances insulin secretion ex vivo, combined deletion of these 2 receptors only minimally affects glucose homeostasis in vivo in mice.


Assuntos
Glucose/metabolismo , Receptores Acoplados a Proteínas G/genética , Animais , Feminino , Deleção de Genes , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Homeostase/genética , Insulina/metabolismo , Secreção de Insulina/genética , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Mol Metab ; 45: 101166, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33484949

RESUMO

OBJECTIVE: Maintenance of glucose homeostasis requires the precise regulation of hormone secretion from the endocrine pancreas. Free fatty acid receptor 4 (FFAR4/GPR120) is a G protein-coupled receptor whose activation in islets of Langerhans promotes insulin and glucagon secretion and inhibits somatostatin secretion. However, the contribution of individual islet cell types (α, ß, and δ cells) to the insulinotropic and glucagonotropic effects of GPR120 remains unclear. As gpr120 mRNA is enriched in somatostatin-secreting δ cells, we hypothesized that GPR120 activation stimulates insulin and glucagon secretion via inhibition of somatostatin release. METHODS: Glucose tolerance tests were performed in mice after administration of selective GPR120 agonist Compound A. Insulin, glucagon, and somatostatin secretion were measured in static incubations of isolated mouse islets in response to endogenous (ω-3 polyunsaturated fatty acids) and/or pharmacological (Compound A and AZ-13581837) GPR120 agonists. The effect of Compound A on hormone secretion was tested further in islets isolated from mice with global or somatostatin cell-specific knock-out of gpr120. Gpr120 expression was assessed in pancreatic sections by RNA in situ hybridization. Cyclic AMP (cAMP) and calcium dynamics in response to pharmacological GPR120 agonists were measured specifically in α, ß, and δ cells in intact islets using cAMPER and GCaMP6 reporter mice, respectively. RESULTS: Acute exposure to Compound A increased glucose tolerance, circulating insulin, and glucagon levels in vivo. Endogenous and/or pharmacological GPR120 agonists reduced somatostatin secretion in isolated islets and concomitantly demonstrated dose-dependent potentiation of glucose-stimulated insulin secretion and arginine-stimulated glucagon secretion. Gpr120 was enriched in δ cells. Pharmacological GPR120 agonists reduced cAMP and calcium levels in δ cells but increased these signals in α and ß cells. Compound A-mediated inhibition of somatostatin secretion was insensitive to pertussis toxin. The effect of Compound A on hormone secretion was completely absent in islets from mice with either global or somatostatin cell-specific deletion of gpr120 and partially reduced upon blockade of somatostatin receptor signaling by cyclosomatostatin. CONCLUSIONS: Inhibitory GPR120 signaling in δ cells contributes to both insulin and glucagon secretion in part by mitigating somatostatin release.


Assuntos
Ácidos Graxos não Esterificados/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Secretoras de Somatostatina/metabolismo , Animais , Feminino , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Teste de Tolerância a Glucose , Homeostase , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Somatostatina/metabolismo
7.
Nat Rev Endocrinol ; 17(3): 162-175, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33495605

RESUMO

Therapeutic approaches to the treatment of type 2 diabetes mellitus that are designed to increase insulin secretion either directly target ß-cells or indirectly target gastrointestinal enteroendocrine cells (EECs), which release hormones that modulate insulin secretion (for example, incretins). Given that ß-cells and EECs both express a large array of G protein-coupled receptors (GPCRs) that modulate insulin secretion, considerable research and development efforts have been undertaken to design therapeutic drugs targeting these GPCRs. Among them are GPCRs specific for free fatty acid ligands (lipid GPCRs), including free fatty acid receptor 1 (FFA1, otherwise known as GPR40), FFA2 (GPR43), FFA3 (GPR41) and FFA4 (GPR120), as well as the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). These lipid GPCRs have demonstrated important roles in the control of islet and gut hormone secretion. Advances in lipid GPCR pharmacology have led to the identification of a number of synthetic agonists that exert beneficial effects on glucose homeostasis in preclinical studies. Yet, translation of these promising results to the clinic has so far been disappointing. In this Review, we present the physiological roles, pharmacology and clinical studies of these lipid receptors and discuss the challenges associated with their clinical development for the treatment of type 2 diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Lipídeos de Membrana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Sistemas de Liberação de Medicamentos/tendências , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/metabolismo
8.
Diabetes ; 69(3): 369-380, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31882563

RESUMO

The molecular mechanisms of ß-cell compensation to metabolic stress are poorly understood. We previously observed that nutrient-induced ß-cell proliferation in rats is dependent on epidermal growth factor receptor (EGFR) signaling. The aim of this study was to determine the role of the EGFR ligand heparin-binding EGF-like growth factor (HB-EGF) in the ß-cell proliferative response to glucose, a ß-cell mitogen and key regulator of ß-cell mass in response to increased insulin demand. We show that exposure of isolated rat and human islets to HB-EGF stimulates ß-cell proliferation. In rat islets, inhibition of EGFR or HB-EGF blocks the proliferative response not only to HB-EGF but also to glucose. Furthermore, knockdown of HB-EGF in rat islets blocks ß-cell proliferation in response to glucose ex vivo and in vivo in transplanted glucose-infused rats. Mechanistically, we demonstrate that HB-EGF mRNA levels are increased in ß-cells in response to glucose in a carbohydrate-response element-binding protein (ChREBP)-dependent manner. In addition, chromatin immunoprecipitation studies identified ChREBP binding sites in proximity to the HB-EGF gene. Finally, inhibition of Src family kinases, known to be involved in HB-EGF processing, abrogated glucose-induced ß-cell proliferation. Our findings identify a novel glucose/HB-EGF/EGFR axis implicated in ß-cell compensation to increased metabolic demand.


Assuntos
Proliferação de Células/genética , Receptores ErbB/metabolismo , Glucose/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Células Secretoras de Insulina/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Receptores ErbB/antagonistas & inibidores , Técnicas de Silenciamento de Genes , Glucose/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais , Quinases da Família src/antagonistas & inibidores
9.
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
10.
Diabetes ; 67(1): 71-77, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29038309

RESUMO

Βß-Cell adaptation to insulin resistance is necessary to maintain glucose homeostasis in obesity. Failure of this mechanism is a hallmark of type 2 diabetes (T2D). Hence, factors controlling functional ß-cell compensation are potentially important targets for the treatment of T2D. Protein kinase D1 (PKD1) integrates diverse signals in the ß-cell and plays a critical role in the control of insulin secretion. However, the role of ß-cell PKD1 in glucose homeostasis in vivo is essentially unknown. Using ß-cell-specific, inducible PKD1 knockout mice (ßPKD1KO), we examined the role of ß-cell PKD1 under basal conditions and during high-fat feeding. ßPKD1KO mice under a chow diet presented no significant difference in glucose tolerance or insulin secretion compared with mice expressing the Cre transgene alone; however, when compared with wild-type mice, both groups developed glucose intolerance. Under a high-fat diet, deletion of PKD1 in ß-cells worsened hyperglycemia, hyperinsulinemia, and glucose intolerance. This was accompanied by impaired glucose-induced insulin secretion both in vivo in hyperglycemic clamps and ex vivo in isolated islets from high-fat diet-fed ßPKD1KO mice without changes in islet mass. This study demonstrates an essential role for PKD1 in the ß-cell adaptive secretory response to high-fat feeding in mice.


Assuntos
Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína Quinase C/metabolismo , Animais , Western Blotting , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Eletroforese em Gel de Poliacrilamida , Feminino , Teste de Tolerância a Glucose , Secreção de Insulina , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase C/genética
11.
Biochimie ; 143: 10-17, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28987628

RESUMO

Excess consumption of energy-dense foods combined with a sedentary lifestyle is driving an obesity epidemic. Although obesity is closely associated with insulin resistance, most individuals meet the insulin demand by increasing their functional ß-cell mass. Those who eventually develop type 2 diabetes are distinguished by a failure in this compensatory process. Although a causal role of insulin resistance in compensatory ß-cell responses has received considerable experimental support, precisely how the ß cell senses changes in the metabolic environment is still unknown. As metabolism of glucose, lipids and amino acids is profoundly altered in obesity, it is not surprising that these nutrients are conspicuous among the factors proposed to contribute. In this review we summarise our understanding of the role of nutrients, in particular glucose, fatty acids and amino acids in ß-cell compensation with a particular emphasis on their relation to insulin resistance-induced factors and their underlying mechanism of action. Finally, we describe the concept of epigenetic programming and review recent studies illustrating how the status of the ß cell epigenome is a product of its nutrient environment, and how metabolic programming of the ß cell contributes to diabetes risk.


Assuntos
Aminoácidos/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/citologia , Metabolismo dos Lipídeos , Aminoácidos/farmacologia , Animais , Proliferação de Células , Epigênese Genética , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Alimentos , Glucose/farmacologia , Humanos , Resistência à Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Transdução de Sinais
12.
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
13.
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
14.
Mol Metab ; 5(10): 988-996, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27689011

RESUMO

OBJECTIVE: G protein-coupled receptor (GPCR) signaling regulates insulin secretion and pancreatic ß cell-proliferation. While much knowledge has been gained regarding how GPCRs are activated in ß cells, less is known about the mechanisms controlling their deactivation. In many cell types, termination of GPCR signaling is controlled by the family of Regulators of G-protein Signaling (RGS). RGS proteins are expressed in most eukaryotic cells and ensure a timely return to the GPCR inactive state upon removal of the stimulus. The aims of this study were i) to determine if RGS16, the most highly enriched RGS protein in ß cells, regulates insulin secretion and ß-cell proliferation and, if so, ii) to elucidate the mechanisms underlying such effects. METHODS: Mouse and human islets were infected with recombinant adenoviruses expressing shRNA or cDNA sequences to knock-down or overexpress RGS16, respectively. 60 h post-infection, insulin secretion and cAMP levels were measured in static incubations in the presence of glucose and various secretagogues. ß-cell proliferation was measured in infected islets after 72 h in the presence of 16.7 mM glucose ± somatostatin and various inhibitors. RESULTS: RGS16 mRNA levels are strongly up-regulated in islets of Langerhans under hyperglycemic conditions in vivo and ex vivo. RGS16 overexpression stimulated glucose-induced insulin secretion in isolated mouse and human islets while, conversely, insulin secretion was impaired following RGS16 knock-down. Insulin secretion was no longer affected by RGS16 knock-down when islets were pre-treated with pertussis toxin to inactivate Gαi/o proteins, or in the presence of a somatostatin receptor antagonist. RGS16 overexpression increased intracellular cAMP levels, and its effects were blocked by an adenylyl cyclase inhibitor. Finally, RGS16 overexpression prevented the inhibitory effect of somatostatin on insulin secretion and ß-cell proliferation. CONCLUSIONS: Our results identify RGS16 as a novel regulator of ß-cell function that coordinately controls insulin secretion and proliferation by limiting the tonic inhibitory signal exerted by δ-cell-derived somatostatin in islets.

15.
Islets ; 8(6): 157-164, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27700527

RESUMO

Free fatty acid receptor 1 (FFA1/GPR40) plays a key role in the potentiation of glucose-stimulated insulin secretion by fatty acids in pancreatic ß cells. We previously demonstrated that GPR40 signaling leads to cortical actin remodeling and potentiates the second phase of insulin secretion. In this study, we examined the role of p21 activated kinase 4 (PAK4), a known regulator of cytoskeletal dynamics, in GPR40-dependent potentiation of insulin secretion. The fatty acid oleate induced PAK4 phosphorylation in human islets, in isolated mouse islets and in the insulin secreting cell line INS832/13. However, oleate-induced PAK4 phosphorylation was not observed in GPR40-null mouse islets. siRNA-mediated knockdown of PAK4 in INS832/13 cells abrogated the potentiation of insulin secretion by oleate, whereas PAK7 knockdown had no effect. Our results indicate that PAK4 plays an important role in the potentiation of insulin secretion by fatty acids downstream of GPR40.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Ácido Oleico/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/metabolismo , Animais , Linhagem Celular , Glucose/farmacologia , Humanos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Knockout , Fosforilação , RNA Interferente Pequeno , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos
16.
J Clin Invest ; 126(9): 3598-612, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27525435

RESUMO

Disorders of glucose homeostasis are common in chronic kidney disease (CKD) and are associated with increased mortality, but the mechanisms of impaired insulin secretion in this disease remain unclear. Here, we tested the hypothesis that defective insulin secretion in CKD is caused by a direct effect of urea on pancreatic ß cells. In a murine model in which CKD is induced by 5/6 nephrectomy (CKD mice), we observed defects in glucose-stimulated insulin secretion in vivo and in isolated islets. Similarly, insulin secretion was impaired in normal mouse and human islets that were cultured with disease-relevant concentrations of urea and in islets from normal mice treated orally with urea for 3 weeks. In CKD mouse islets as well as urea-exposed normal islets, we observed an increase in oxidative stress and protein O-GlcNAcylation. Protein O-GlcNAcylation was also observed in pancreatic sections from CKD patients. Impairment of insulin secretion in both CKD mouse and urea-exposed islets was associated with reduced glucose utilization and activity of phosphofructokinase 1 (PFK-1), which could be reversed by inhibiting O-GlcNAcylation. Inhibition of O-GlcNAcylation also restored insulin secretion in both mouse models. These results suggest that insulin secretory defects associated with CKD arise from elevated circulating levels of urea that increase islet protein O-GlcNAcylation and impair glycolysis.


Assuntos
Glicólise , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Falência Renal Crônica/metabolismo , Ureia/química , Animais , Antioxidantes/metabolismo , Cianatos/química , Modelos Animais de Doenças , Exocitose , Glucoquinase/metabolismo , Glucose/metabolismo , Teste de Tolerância a Glucose , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Fosfofrutoquinase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Uremia/metabolismo
17.
Endocrinology ; 157(3): 1299-306, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26671180

RESUMO

Mouse ß-cell-specific reporter lines have played a key role in diabetes research. Although the rat provides several advantages, its use has lagged behind the mouse due to the relative paucity of genetic models. In this report we describe the generation and characterization of transgenic rats expressing a Renilla luciferase (RLuc)-enhanced yellow fluorescent protein (YFP) fusion under control of a 9-kb genomic fragment from the rat ins2 gene (RIP7-RLuc-YFP). Analysis of RLuc luminescence and YFP fluorescence revealed that reporter expression is restricted to ß-cells in the adult rat. Physiological characteristics including body weight, fat and lean mass, fasting and fed glucose levels, glucose and insulin tolerance, and ß-cell mass were similar between two RIP7-RLuc-YFP lines and wild-type littermates. Glucose-induced insulin secretion in isolated islets was indistinguishable from controls in one of the lines, whereas surprisingly, insulin secretion was defective in the second line. Consequently, subsequent studies were limited to the former line. We asked whether transgene activity was responsive to glucose as shown previously for the ins2 gene. Exposing islets ex vivo to high glucose (16.7 mM) or in vivo infusion of glucose for 24 hours increased luciferase activity in islets, whereas the fraction of YFP-positive ß-cells after glucose infusion was unchanged. Finally, we showed that fluorescence-activated cell sorting of YFP-positive islet cells can be used to enrich for ß-cells. Overall, this transgenic line will enable for the first time the application of both fluorescence and bioluminescence/luminescence-based approaches for the study of rat ß-cells.


Assuntos
Proteínas de Bactérias/genética , Genes Reporter/genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Luciferases de Renilla/genética , Proteínas Luminescentes/genética , Modelos Animais , Animais , Fusão Gênica Artificial , Proteínas de Bactérias/efeitos dos fármacos , Glicemia/metabolismo , Citometria de Fluxo , Genes Reporter/efeitos dos fármacos , Glucose/farmacologia , Teste de Tolerância a Glucose , Células Secretoras de Insulina/efeitos dos fármacos , Luciferases de Renilla/efeitos dos fármacos , Proteínas Luminescentes/efeitos dos fármacos , Masculino , Regiões Promotoras Genéticas , Ratos , Ratos Transgênicos
18.
Diabetes ; 64(12): 4112-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26283735

RESUMO

Cystic fibrosis (CF) is the result of mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). CF-related diabetes affects 50% of adult CF patients. How CFTR deficiency predisposes to diabetes is unknown. Herein, we examined the impact of the most frequent cftr mutation in humans, deletion of phenylalanine at position 508 (ΔF508), on glucose homeostasis in mice. We compared ΔF508 mutant mice with wild-type (WT) littermates. Twelve-week-old male ΔF508 mutants had lower body weight, improved oral glucose tolerance, and a trend toward higher insulin tolerance. Glucose-induced insulin secretion was slightly diminished in ΔF508 mutant islets, due to reduced insulin content, but ΔF508 mutant islets were not more sensitive to proinflammatory cytokines than WT islets. Hyperglycemic clamps confirmed an increase in insulin sensitivity with normal ß-cell function in 12- and 18-week-old ΔF508 mutants. In contrast, 24-week-old ΔF508 mutants exhibited insulin resistance and reduced ß-cell function. ß-Cell mass was unaffected at 11 weeks of age but was significantly lower in ΔF508 mutants versus controls at 24 weeks. This was not associated with gross pancreatic pathology. We conclude that the ΔF508 CFTR mutation does not lead to an intrinsic ß-cell secretory defect but is associated with insulin resistance and a ß-cell mass deficit in aging mutants.


Assuntos
Envelhecimento , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mutação , Animais , Cruzamentos Genéticos , Fibrose Cística/genética , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diabetes Mellitus/etiologia , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Insulina/sangue , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos Endogâmicos , Camundongos Mutantes , Técnicas de Cultura de Tecidos
19.
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
20.
Islets ; 6(4): e982376, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25437380

RESUMO

The transcription factor Pancreatic and Duodenal Homeobox-1 (PDX-1) plays a major role in the development and function of pancreatic ß-cells and its mutation results in diabetes. In adult ß-cells, glucose stimulates transcription of the insulin gene in part by regulating PDX-1 expression, stability and activity. Glucose is also thought to modulate PDX-1 nuclear translocation but in vitro studies examining nucleo-cytoplasmic shuttling of endogenous or ectopically expressed PDX-1 in insulin-secreting cell lines have led to conflicting results. Here we show that endogenous PDX-1 undergoes translocation from the cytoplasm to the nucleus in response to glucose in dispersed rat islets but not in insulin-secreting MIN6, HIT-T15, or INS832/13 cells. Interestingly, however, we found that a PDX-1-GFP fusion protein can shuttle from the cytoplasm to the nucleus in response to glucose stimulation in HIT-T15 cells. Our results suggest that the regulation of endogenous PDX-1 sub-cellular localization by glucose is observed in primary islets and that care should be taken when interpreting data from insulin-secreting cell lines.


Assuntos
Glucose/farmacologia , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Imuno-Histoquímica , Masculino , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...