Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 396
Filtrar
1.
EMBO J ; 42(23): e114086, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37807855

RESUMO

The immune response is an energy-demanding process that must be coordinated with systemic metabolic changes redirecting nutrients from stores to the immune system. Although this interplay is fundamental for the function of the immune system, the underlying mechanisms remain elusive. Our data show that the pro-inflammatory polarization of Drosophila macrophages is coupled to the production of the insulin antagonist ImpL2 through the activity of the transcription factor HIF1α. ImpL2 production, reflecting nutritional demands of activated macrophages, subsequently impairs insulin signaling in the fat body, thereby triggering FOXO-driven mobilization of lipoproteins. This metabolic adaptation is fundamental for the function of the immune system and an individual's resistance to infection. We demonstrated that analogically to Drosophila, mammalian immune-activated macrophages produce ImpL2 homolog IGFBP7 in a HIF1α-dependent manner and that enhanced IGFBP7 production by these cells induces mobilization of lipoproteins from hepatocytes. Hence, the production of ImpL2/IGFBP7 by macrophages represents an evolutionarily conserved mechanism by which macrophages alleviate insulin signaling in the central metabolic organ to secure nutrients necessary for their function upon bacterial infection.


Assuntos
Infecções Bacterianas , Proteínas de Drosophila , Resistência à Insulina , Animais , Antagonistas da Insulina/metabolismo , Antagonistas da Insulina/farmacologia , Drosophila/metabolismo , Insulina/metabolismo , Macrófagos/metabolismo , Infecções Bacterianas/metabolismo , Mamíferos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Proteínas de Drosophila/metabolismo
3.
ACS Chem Neurosci ; 13(13): 2006-2016, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35704461

RESUMO

Substoichiometric aggregation inhibition of human islet amyloid polypeptide (IAPP), the hallmark of type 2 diabetes impacting millions of people, is crucial for developing clinic therapies, yet it remains challenging given that many candidate inhibitors require high doses. Intriguingly, insulin, the key regulatory polypeptide on blood glucose levels that are cosynthesized, costored, and cosecreted with IAPP by pancreatic ß cells, has been identified as a potent inhibitor that can suppress IAPP amyloid aggregation at substoichiometric concentrations. Here, we computationally investigated the molecular mechanisms of the substoichiometric inhibition of insulin against the aggregation of IAPP and the incompletely processed IAPP (proIAPP) using discrete molecular dynamics simulations. Our results suggest that the amyloid aggregations of both IAPP and proIAPP might be disrupted by insulin through its binding with the shared amyloidogenic core sequences. However, the N-terminus of proIAPP competed with the amyloidogenic core sequences for the insulin interactions, resulting in attenuated inhibition by insulin. Moreover, insulin preferred to bind the elongation surfaces of IAPP seeds with fibril-like structure, with a stronger affinity than that of IAPP monomers. The capping of elongation surfaces by a small amount of insulin sterically prohibited the seed growth via monomer addition, achieving the substoichiometric inhibition. Together, our computational results provided molecular insights for the substoichiometric inhibition of insulin against IAPP aggregation, also the weakened effect on proIAPP. The uncovered substoichiometric inhibition by capping the elongation of amyloid seeds or fibrils may guide the rational designs of new potent inhibitors effective at low doses.


Assuntos
Amiloidose , Diabetes Mellitus Tipo 2 , Insulina , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Amiloide/metabolismo , Amiloidose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Antagonistas da Insulina , Polipeptídeo Amiloide das Ilhotas Pancreáticas/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo
4.
Nature ; 590(7845): 326-331, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33505018

RESUMO

Resistance to insulin and insulin-like growth factor 1 (IGF1) in pancreatic ß-cells causes overt diabetes in mice; thus, therapies that sensitize ß-cells to insulin may protect patients with diabetes against ß-cell failure1-3. Here we identify an inhibitor of insulin receptor (INSR) and IGF1 receptor (IGF1R) signalling in mouse ß-cells, which we name the insulin inhibitory receptor (inceptor; encoded by the gene Iir). Inceptor contains an extracellular cysteine-rich domain with similarities to INSR and IGF1R4, and a mannose 6-phosphate receptor domain that is also found in the IGF2 receptor (IGF2R)5. Knockout mice that lack inceptor (Iir-/-) exhibit signs of hyperinsulinaemia and hypoglycaemia, and die within a few hours of birth. Molecular and cellular analyses of embryonic and postnatal pancreases from Iir-/- mice showed an increase in the activation of INSR-IGF1R in Iir-/- pancreatic tissue, resulting in an increase in the proliferation and mass of ß-cells. Similarly, inducible ß-cell-specific Iir-/- knockout in adult mice and in ex vivo islets led to an increase in the activation of INSR-IGF1R and increased proliferation of ß-cells, resulting in improved glucose tolerance in vivo. Mechanistically, inceptor interacts with INSR-IGF1R to facilitate clathrin-mediated endocytosis for receptor desensitization. Blocking this physical interaction using monoclonal antibodies against the extracellular domain of inceptor resulted in the retention of inceptor and INSR at the plasma membrane to sustain the activation of INSR-IGF1R in ß-cells. Together, our findings show that inceptor shields insulin-producing ß-cells from constitutive pathway activation, and identify inceptor as a potential molecular target for INSR-IGF1R sensitization and diabetes therapy.


Assuntos
Glicemia/metabolismo , Antagonistas da Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Animais , Glicemia/análise , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Tamanho Celular , Clatrina/metabolismo , Células Endócrinas/metabolismo , Endocitose , Retículo Endoplasmático/metabolismo , Teste de Tolerância a Glucose , Complexo de Golgi/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Proteínas de Membrana , Camundongos , Proteínas de Neoplasias/química , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia
5.
Pharmacol Res ; 165: 105417, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33401003

RESUMO

Carrageenan (CGN) is a common food additive, and questions have been raised regarding its safety for human consumption. The purpose of this study was to investigate the impact of κ-CGN on glucose intolerance and insulin resistance from the perspective that κ-CGN may interfere with insulin receptor function and affect insulin sensitivity and signaling, thereby leading to body weight loss. The health effects of κ-CGN on C57BL/6 mice were assessed over a 90-d period by monitoring changes in body weight, glucose tolerance, insulin tolerance, fasting glucose and insulin levels, and expression of insulin-pathway-related proteins. Furthermore, HepG2 cells were used to detect the binding of κ-CGN on insulin receptor and measure its effect on downstream signal transduction. In mice, κ-CGN treatment reduced weight gain without affecting food intake. Glucose and insulin tolerance tests revealed that κ-CGN treatment increased blood glucose levels and glycosylated hemoglobin levels, while hepatic and muscle glycogen levels were decreased, suggesting that κ-CGN affected glucose metabolism in mice. Interestingly, κ-CGN treatment did not cause typical diabetic symptoms in mice, as indicated by low levels of fasting and postprandial blood glucose, in addition to normal pancreatic tissue and insulin secretion. The binding studies revealed that κ-CGN could competitively bind to the insulin receptor with FITC-insulin and thereby disrupt PI3K and Akt activation, thus suppressing expression of glucose transporters and glycogen synthase. In summary, this study revealed that κ-CGN reduced weight gain without affecting food intake, but impaired glucose metabolism in mice by interfering with insulin binding to receptors, thereby affecting the sensitivity of insulin and inhibiting the insulin PI3K/AKT signaling pathway, causing non-diabetic weight gain reduction.


Assuntos
Carragenina/efeitos adversos , Antagonistas da Insulina/efeitos adversos , Insulina/metabolismo , Doenças Metabólicas/induzido quimicamente , Animais , Western Blotting , Citometria de Fluxo , Glucose/metabolismo , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Aumento de Peso/efeitos dos fármacos
6.
J Physiol Pharmacol ; 71(1)2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32554844

RESUMO

Adropin is a peptide hormone which is produced in brain and peripheral tissues such as liver. It was found that adropin modulates lipid and glucose homeostasis by interacting with hepatocytes and myocytes. Adropin enhances insulin sensitivity and alleviates hyperinsulinemia in animal models with high-fat diet-induced insulin resistance. However, it is unknown whether adropin regulates insulin secretion and proliferation of beta cells. Therefore, we studied the effects of adropin on insulin secretion in INS-1E cells as well as isolated pancreatic islets. Furthermore, we assessed the influence of adropin on insulin mRNA expression, cell viability and proliferation in INS-1E cells. Pancreatic islets were isolated from male Wistar rats. mRNA expression was evaluated using real-time PCR and cell viability by MTT assay. Cell replication was measured by BrdU incorporation and insulin secretion by RIA. We found that adropin suppresses insulin mRNA expression in INS-1E cells. Moreover, adropin attenuates glucose-induced insulin secretion in INS-1E cells as well as in isolated pancreatic islets. In addition, using INS-1E cells we found that adropin suppresses glucose-induced cAMP production. However, adropin fails to modulate INS-1E cell viability and proliferation. In summary, we found adropin suppresses insulin mRNA expression and secretion, without affecting beta cell viability or proliferation.


Assuntos
Proteínas Sanguíneas/farmacologia , Antagonistas da Insulina/farmacologia , Secreção de Insulina/fisiologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Peptídeos/farmacologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Ratos , Ratos Wistar
7.
Toxicol Sci ; 175(2): 149-155, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32207534

RESUMO

In 2014, it was estimated that more than 1.9 billion adults were overweight with over 600 million classifiable as obese. Approximately two-thirds of U.S. adults over 20 years of age are currently overweight with about 35% classified as obese, a figure thought likely to reach 42% by 2030 in those over 18 years of age. Adipose cells from stored body fat secrete estrogen and a very large number (> 500) of biologically active substances termed adipokines, in addition to inducing, by other cell-driven effects, pathological alterations in insulin pathways. The U.S. National Cancer Institute reports that exposure to the hormone disrupting and proinflammatory effects of excess adipose tissue are associated with an increased risk for 11 different cancers. Obesity is also associated with a number of serious non-neoplastic conditions including metabolic syndrome and type 2 diabetes; menstrual cycle irregularities and lowered fertility (men and women); and abnormal bone morphology in a subset of female patients. In men hypogonadism, low testosterone levels, benign prostatic hyperplasia, and lowered sperm counts have been reported. In developed countries, the endogenous adverse health burden associated with obesity is only matched, quantitatively and qualitatively, by the exogenous toxicity of cigarette smoking. The investigation of possible hormonal and/or proinflammatory effects of chemicals should include an assessment of the profound endocrine alterations associated with obesity.


Assuntos
Tecido Adiposo/fisiopatologia , Doença Crônica , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Síndrome Metabólica/complicações , Obesidade/complicações , Obesidade/fisiopatologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Diabetes Mellitus Tipo 2/epidemiologia , Feminino , Humanos , Antagonistas da Insulina , Masculino , Síndrome Metabólica/epidemiologia , Síndrome Metabólica/fisiopatologia , Pessoa de Meia-Idade , Obesidade/epidemiologia , Estados Unidos/epidemiologia , Adulto Jovem
8.
Animal ; 13(12): 2840-2846, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31148536

RESUMO

Consumers' demand of leaner meat products is a challenge. Although betaine and conjugated linoleic acid (CLA) have the potential to decrease porcine adipose tissue, their mode of action is poorly understood. The aim of the study was to determine the lipolytic effect of betaine and CLA in the adipose tissue of Iberian pigs. Adipose tissue explants from five pigs (38 kg BW) were prepared from dorsal subcutaneous adipose tissue samples and cultivated for 2 h (acute experiments) or 72 h (chronic experiments). Treatments included 100 µM linoleic acid (control), 100 µM trans-10, cis-12 CLA, 100 µM linoleic acid + 1 mM betaine and 100 µM trans-10, cis-12 CLA + 1 mM betaine (CLABET). To examine the ability of betaine or CLA to inhibit insulin's suppression of isoproterenol-stimulated lipolysis, test medium was amended with 1 µM isoproterenol ±10 nM insulin. Media glycerol was measured at the end of the incubations. Acute lipolysis (2 h) was increased by CLA and CLABET (85% to 121%; P < 0.05) under basal conditions. When lipolysis was stimulated with isoproterenol (1090%), acute exposure to betaine tended to increase (13%; P = 0.071), while CLA and CLABET increased (14% to 18%; P < 0.05) isoproterenol-stimulated lipolysis compared with control. When insulin was added to isoproterenol-stimulated explants, lipolytic rate was decreased by 50% (P < 0.001). However, supplementation of betaine to the insulin + isoproterenol-containing medium tended to increase (P = 0.07), while CLABET increased (45%; P < 0.05) lipolysis, partly counteracting insulin inhibition. When culture was extended for 72 h, CLA decreased lipolysis under basal conditions (18%; P < 0.05) with no effect of betaine and CLABET (P > 0.10). When lipolysis was stimulated by isoproterenol (125% increase in rate compared with basal), CLA and CLABET decreased glycerol release (27%; P < 0.001) compared with control (isoproterenol alone). When insulin was added to isoproterenol-stimulated explants, isoproterenol stimulation of lipolysis was completely blunted and neither betaine nor CLA altered the inhibitory effect of insulin on lipolysis. Isoproterenol, and especially isoproterenol + insulin, stimulated leptin secretion compared with basal conditions (68% and 464%, respectively; P < 0.001), with no effect of CLA or betaine (P > 0.10). CLA decreased leptin release (25%; P < 0.001) when insulin was present in the media, partially inhibiting insulin stimulation of leptin release. In conclusion, betaine and CLA produced a biphasic response regarding lipolysis so that glycerol release was increased in acute conditions, while CLA decreased glycerol release and betaine had no effect in chronic conditions. Furthermore, CLA and CLABET indirectly increased lipolysis by reducing insulin-mediated inhibition of lipolysis during acute conditions.


Assuntos
Betaína/farmacologia , Antagonistas da Insulina/farmacologia , Insulina/metabolismo , Ácidos Linoleicos Conjugados/farmacologia , Lipólise/efeitos dos fármacos , Suínos/fisiologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Glicerol/metabolismo , Isoproterenol/metabolismo , Leptina/metabolismo , Masculino
9.
Am J Physiol Endocrinol Metab ; 317(2): E250-E260, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31084489

RESUMO

Fetuin-A (Fet-A), a hepatokine associated with insulin resistance, obesity, and incident type 2 diabetes, is shown to exist in both phosphorylated and dephosphorylated forms in circulation. However, studies on fetuin-A phosphorylation status in insulin-resistant conditions and its functional significance are limited. We demonstrate that serum phosphofetuin-A (Ser312) levels were significantly elevated in high-fat diet-induced obese mice, insulin-resistant Zucker diabetic fatty rats, and in individuals with obesity who are insulin resistant. Unlike serum total fetuin-A, serum phosphofetuin-A was associated with body weight, insulin, and markers of insulin resistance. To characterize potential mechanisms, fetuin-A was purified from Hep3B human hepatoma cells. Hep3B Fet-A was phosphorylated (Ser312) and inhibited insulin-stimulated glucose uptake and glycogen synthesis in L6GLUT4 myoblasts. Furthermore, single (Ser312Ala) and double (Ser312Ala + Ser120Ala) phosphorylation-defective Fet-A mutants were without effect on glucose uptake and glycogen synthesis in L6GLUT4 myoblasts. Together, our studies demonstrate that phosphorylation status of Fet-A (Ser312) is associated with obesity and insulin resistance and raise the possibility that Fet-A phosphorylation may play a role in regulation of insulin action.


Assuntos
Resistência à Insulina/fisiologia , Obesidade/metabolismo , Proteínas Quinases/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Células 3T3-L1 , Adulto , Idoso , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Insulina/metabolismo , Antagonistas da Insulina/metabolismo , Antagonistas da Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fosforilação , Ratos , Ratos Zucker , alfa-2-Glicoproteína-HS/farmacologia
10.
Molecules ; 23(11)2018 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-30380710

RESUMO

In Mexico one in 14 deaths are caused by diabetes mellitus (DM) or by the macro and microvascular disorders derived from it. A continuous hyperglycemic state is characteristic of DM, resulting from a sustained state of insulin resistance and/or a dysfunction of ß-pancreatic cells. Acaciella angustissima is a little studied species showing a significant antioxidant activity that can be used as treatment of this disease or preventive against the complications. The objective of this study was to explore the effect of oral administration of A. angustissima methanol extract on physiological parameters of streptozotocin-induced diabetic rats. The results indicated a significant reduction in blood glucose levels, an increase in serum insulin concentration, a decrease in lipid levels and an improvement in the parameters of kidney damage by applying a concentration of 100 mg/Kg B.W. However, glucose uptake activity was not observed in the adipocyte assay. Moreover, the extract of A. angustissima displayed potential for the complementary treatment of diabetes and its complications likely due to the presence of bioactive compounds such as protocatechuic acid. This study demonstrated that methanol extract of Acacciella angustissima has an antidiabetic effect by reducing the levels of glucose, insulin and improved physiological parameters, hypolipidemic effect, oxidative stress and renal damage in diabetic rats.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Fabaceae/química , Hipolipemiantes/administração & dosagem , Extratos Vegetais/administração & dosagem , Animais , Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Frutas/química , Humanos , Hipolipemiantes/química , Insulina/sangue , Antagonistas da Insulina/administração & dosagem , Antagonistas da Insulina/química , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/química , Ratos
11.
J Dairy Sci ; 101(4): 3428-3432, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29395144

RESUMO

In nonruminants, the sphingolipid ceramide inhibits insulin sensitivity by inactivating protein kinase B (AKT) within the insulin-signaling pathway. We have established that ceramide accrual develops with impaired systemic insulin action in ruminants during the transition from gestation to lactation, dietary palmitic acid supplementation, or controlled nutrient restriction. We hypothesized that ceramide promotes AKT inactivation and antagonizes insulin sensitivity in primary bovine adipocytes. Stromal-vascular cells were grown from bovine adipose tissue explants and cultured in differentiation media. To modify ceramide supply, we treated differentiated adipocytes with (1) myriocin, an inhibitor of de novo ceramide synthesis, or (2) cell-permeable C2:0-ceramide. Insulin-stimulated AKT activation (i.e., phosphorylation) and 2-deoxy-D-[3H]-glucose (2DOG) uptake were measured. Treatment of adipocytes with myriocin consistently decreased concentrations of ceramide, monohexosylceramide, and lactosylceramide. The insulin-stimulated ratio of phosphorylated AKT to total AKT was increased with myriocin but decreased with C2:0-ceramide. Moreover, adipocyte insulin-stimulated 2DOG uptake was decreased with C2:0-ceramide and increased with myriocin. We conclude that ceramide inhibits insulin-stimulated glucose uptake by downregulating AKT activation in primary bovine adipocytes.


Assuntos
Adipócitos/metabolismo , Ceramidas/farmacologia , Antagonistas da Insulina/farmacologia , Resistência à Insulina , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Bovinos , Masculino
12.
Peptides ; 100: 18-23, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29412818

RESUMO

In the treatment of insulin-dependent diabetes the risk of a fatal insulin overdose is a persistent fear to most patients. In order to potentially reduce the risk of overdose, we report the design, synthesis, and biochemical characterization of a set of insulin analogs designed to be fractionally reduced in maximal agonism at the insulin receptor isoforms. These analogs consist of native insulin that is site-specifically conjugated to a peptide-based insulin receptor antagonist. The structural refinement of the antagonist once conjugated to insulin provided a set of partial agonists exhibiting between 25 and 70% of the maximal agonism of native insulin at the two insulin receptor isoforms, with only slight differences in inherent potency. These rationally-designed partial agonists provide an approach to interrogate whether control of maximal activity can provide glycemic control with reduced hypoglycemic risk.


Assuntos
Antígenos CD/química , Diabetes Mellitus Tipo 1/tratamento farmacológico , Overdose de Drogas/prevenção & controle , Insulina/uso terapêutico , Peptídeos/uso terapêutico , Receptor de Insulina/química , Antígenos CD/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Overdose de Drogas/genética , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/uso terapêutico , Insulina/efeitos adversos , Insulina/análogos & derivados , Insulina/química , Antagonistas da Insulina/química , Antagonistas da Insulina/uso terapêutico , Peptídeos/química , Isoformas de Proteínas/química , Proteólise/efeitos dos fármacos , Receptor de Insulina/genética , Transdução de Sinais/efeitos dos fármacos
13.
Braz. J. Pharm. Sci. (Online) ; 54(1): e17192, 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-951911

RESUMO

ABSTRACT In clinical practice, simvastatin is usually used in the treatment of dyslipidemia patients and those at risk of or with established cardiovascular disease. However, previous studies have shown that simvastatin has the potential to affect glycemic parameters as it reportedly reduced insulin secretion and sensitivity. The exact mechanism by which simvastatin affects glycemia is still unknown, but previous studies have postulated the involvement of the glucose-insulin secretion mechanism. This review focuses on the effects of simvastatin, either alone or in combination with other lipid lowering agents, antidiabetics and antihypertensives, on glucose homeostasis. Some studies have reported that simvastatin might impair the levels of glucose metabolism markers in the blood while others have reported no effect or improvement in glycemia.


Assuntos
Sinvastatina/efeitos adversos , Interações Medicamentosas , Glucose/efeitos adversos , Antagonistas da Insulina , Técnicas In Vitro/instrumentação , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Transportador de Glucose Tipo 2
14.
Neurochem Int ; 110: 75-83, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28919254

RESUMO

The high consumption of saturated lipids has been largely associated with the increasing prevalence of metabolic diseases. In particular, saturated fatty acids such as palmitic acid (PA) have been implicated in the development of insulin resistance in peripheral tissues. However, how neurons develop insulin resistance in response to lipid overload is not fully understood. Here, we used cultured rat cortical neurons and differentiated human neuroblastoma cells to demonstrate that PA blocks insulin-induced metabolic activation, inhibits the activation of the insulin/PI3K/Akt pathway and activates mTOR kinase downstream of Akt. Despite the fact that fatty acids are not normally used as a significant source of fuel by neural cells, we also found that short-term neuronal exposure to PA reduces the NAD+/NADH ratio, indicating that PA modifies the neuronal energy balance. Finally, inhibiting mitochondrial ROS production with mitoTEMPO prevented the deleterious effect of PA on insulin signaling. This work provides novel evidence of the mechanisms behind saturated fatty acid-induced insulin resistance and its metabolic consequences on neuronal cells.


Assuntos
Insulina/metabolismo , Ácido Palmítico/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Humanos , Antagonistas da Insulina/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroblastoma/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Transdução de Sinais
15.
JCI Insight ; 2(17)2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28878130

RESUMO

Human endogenous retroviruses (HERVs), remnants of ancestral viral genomic insertions, are known to represent 8% of the human genome and are associated with several pathologies. In particular, the envelope protein of HERV-W family (HERV-W-Env) has been involved in multiple sclerosis pathogenesis. Investigations to detect HERV-W-Env in a few other autoimmune diseases were negative, except in type-1 diabetes (T1D). In patients suffering from T1D, HERV-W-Env protein was detected in 70% of sera, and its corresponding RNA was detected in 57% of peripheral blood mononuclear cells. While studies on human Langerhans islets evidenced the inhibition of insulin secretion by HERV-W-Env, this endogenous protein was found to be expressed by acinar cells in 75% of human T1D pancreata. An extensive immunohistological analysis further revealed a significant correlation between HERV-W-Env expression and macrophage infiltrates in the exocrine part of human pancreata. Such findings were corroborated by in vivo studies on transgenic mice expressing HERV-W-env gene, which displayed hyperglycemia and decreased levels of insulin, along with immune cell infiltrates in their pancreas. Altogether, these results strongly suggest an involvement of HERV-W-Env in T1D pathogenesis. They also provide potentially novel therapeutic perspectives, since unveiling a pathogenic target in T1D.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/virologia , Retrovirus Endógenos/efeitos dos fármacos , Proteínas do Envelope Viral/fisiologia , Animais , Antivirais/uso terapêutico , Estudos de Coortes , Diabetes Mellitus Tipo 1/complicações , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidade , Feminino , Humanos , Hiperglicemia/complicações , Insulina/metabolismo , Antagonistas da Insulina/farmacologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , RNA Viral/sangue , Proteínas do Envelope Viral/efeitos dos fármacos
16.
Pediatr Cardiol ; 38(6): 1247-1250, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28642988

RESUMO

Congenital hyperinsulinism (CHI) is the most common cause of persistent hypoglycemia in infancy. The mainstay of medical management for CHI is diazoxide. Diazoxide inhibits insulin release from the pancreas, but also causes smooth muscle relaxation and fluid retention so it is typically given with chlorothiazide. In July 2015, the FDA issued a drug safety communication warning that pulmonary hypertension (PH) had been reported in 11 infants being treated with diazoxide and that the PH resolved with withdrawal of diazoxide. All three of the cases in our hospital were admitted to the neonatal intensive care unit (NICU) for hypoglycemia. All patients received thorough radiologic and laboratory evaluations related to their diagnosis of CHI. All initially improved when diazoxide was initiated. Case 1 and case 3 were discharged from the NICU on diazoxide and chlorothiazide. Case 2 developed pulmonary hypertension while still in the NICU days after an increase in diazoxide dosing. Case 1 presented to the emergency room in respiratory distress shortly after discharge from the NICU with evidence of PH and heart failure. Case 3 presented to the emergency room after 2 weeks at home due to a home blood glucose reading that was low and developed PH and heart failure while an inpatient. Discontinuation of diazoxide led to resolution of all three patients' PH within approximately one week. The experience of our hospital indicates that pulmonary hypertension may be more common than previously thought in infants taking diazoxide. It is unclear if these symptoms develop slowly over time or if there is some other, as yet undescribed, trigger for the pulmonary hypertension. Our hospital's experience adds to the body of evidence and suggests these infants may benefit from more surveillance with echocardiography.


Assuntos
Hiperinsulinismo Congênito/tratamento farmacológico , Diazóxido/efeitos adversos , Hipertensão Pulmonar/induzido quimicamente , Antagonistas da Insulina/efeitos adversos , Diazóxido/uso terapêutico , Humanos , Hipertensão Pulmonar/diagnóstico , Recém-Nascido , Antagonistas da Insulina/uso terapêutico , Masculino
17.
Sci Rep ; 7(1): 3156, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28600547

RESUMO

Congenital hyperinsulinism (CHI) is a rare genetic disorder characterized by excess insulin secretion, which results in hypoglycemia. Mutation of sulfonylurea receptor 1 (SUR1), encoded by the ABCC8 gene, is the main cause of CHI. Here, we captured the phenotype of excess insulin secretion through pancreatic differentiation of ABCC8-deficient stem cells generated by the CRISPR/Cas9 system. ABCC8-deficient insulin-producing cells secreted higher insulin than their wild-type counterparts, and the excess insulin secretion was rescued by nifedipine, octreotide and nicorandil. Further, we tested the role of SUR1 in response to different potassium levels and found that dysfunction of SUR1 decreased the insulin secretion rate in low and high potassium environments. Hence, pancreatic differentiation of ABCC8-deficient cells recapitulated the CHI disease phenotype in vitro, which represents an attractive model to further elucidate the function of SUR1 and to develop and screen for novel therapeutic drugs.


Assuntos
Sistemas CRISPR-Cas , Células-Tronco Embrionárias Humanas/metabolismo , Células Secretoras de Insulina/metabolismo , Modelos Biológicos , Receptores de Sulfonilureias/genética , Peptídeo C/antagonistas & inibidores , Peptídeo C/biossíntese , Diferenciação Celular , Hiperinsulinismo Congênito/genética , Hiperinsulinismo Congênito/metabolismo , Hiperinsulinismo Congênito/patologia , Fármacos Gastrointestinais/farmacologia , Edição de Genes/métodos , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Insulina/biossíntese , Antagonistas da Insulina/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Nicorandil/farmacologia , Nifedipino/farmacologia , Octreotida/farmacologia , Fenótipo , Cloreto de Potássio/farmacologia , Receptores de Sulfonilureias/deficiência , Vasodilatadores/farmacologia
18.
J Formos Med Assoc ; 116(2): 80-89, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27131512

RESUMO

BACKGROUND/PURPOSE: Evidence on clinical effectiveness of metformin in ethnic Chinese women with polycystic ovary syndrome (PCOS) remains scarce. Standard diagnostic approaches to identify insulin resistance (IR) cases in PCOS patients might be invasive, labor intensive, and stressful for patients (i.e., euglycemic clamp), or somewhat complicated for clinicians to calculate and monitor in routine practice [i.e., the homeostatic model assessment (HOMA) and quantitative insulin sensitivity check index (QUICKI)]. The aim of this study was to evaluate the clinical effects of metformin in Taiwanese women with PCOS and identify the feasible diagnostic measures of IR for Taiwanese women with PCOS. METHODS: A total of 114 women from a medical center in Taiwan were studied. All were aged between 18 years and 45 years, diagnosed with PCOS according to the Rotterdam criteria, and treated with metformin. Outcome end points were body mass index (BMI) and 2-hour postload glucose and insulin levels from a 75-g oral glucose tolerance test. RESULTS: BMI in overweight patients were significantly improved with metformin treatment duration (p < 0.001). The 2-hour insulin level statistically improved after treatment (before: 80.7 ± 63.9 µIU/mL vs. after: 65.0 ± 60.4 µIU/mL; p = 0.009). The improved 2-hour insulin level was significantly greater in IR patients than in non-IR patients. Compared with the 2-hour postload insulin level, the fasting insulin level provided 18.15% sensitivity and 94.12% specificity, the HOMA yielded 40% sensitivity and 70.58% specificity, and the QUICKI achieved 63.63% sensitivity and 11.76% specificity. CONCLUSION: Clinical outcomes in Taiwanese PCOS women were improved with metformin treatment, especially in overweight and IR patients. The 2-hour postload insulin level appears to be a convenient tool for screening IR in Taiwanese patients.


Assuntos
Antagonistas da Insulina/administração & dosagem , Resistência à Insulina , Metformina/administração & dosagem , Sobrepeso/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Adulto , Glicemia/efeitos dos fármacos , Índice de Massa Corporal , Feminino , Teste de Tolerância a Glucose , Humanos , Uso Off-Label , Síndrome do Ovário Policístico/sangue , Estudos Retrospectivos , Taiwan , Resultado do Tratamento , Adulto Jovem
19.
Metab Syndr Relat Disord ; 14(8): 391-396, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27028341

RESUMO

BACKGROUND: Insulin resistance in women with polycystic ovary syndrome (PCOS) may be mediated, in part, by a deficiency in the insulin-stimulated release of a d-chiro-inositol-inositolphosphoglycan (DCI-IPG) mediator of insulin action. Supporting this idea, several studies have reported improved insulin sensitivity in both lean and obese women with PCOS after oral administration of DCI. Pioglitazone improves insulin sensitivity in women with PCOS, but it is unknown whether this may be contributed by enhanced insulin-stimulated release of the DCI-IPG second messenger. The study aimed to determine if pioglitazone increases release of biologically active DCI-IPG per unit insulin released during an oral glucose tolerance test (OGTT). METHODS: A randomized, double-blind placebo-controlled trial was conducted in 32 women with PCOS at a tertiary referral center in Venezuela. The intervention comprised administration of pioglitazone 45 mg daily or matched placebo for 6 months. Outcome measures included area under curves (AUC) of DCI-IPG (AUCDCI-IPG), insulin (AUCinsulin), and the ratio of AUCDCI-IPG to AUCinsulin during a 2-hr OGTT. RESULTS: After treatment with pioglitazone, AUCinsulin during the OGTT decreased and whole body insulin sensitivity, as determined by the Matsuda index, increased significantly only in the pioglitazone group. The ratio of AUCDCI-IPG/AUCinsulin increased in the pioglitazone group by 1.85-fold (P < 0.0001) with no significant change in the placebo group. Change in Matsuda index correlated with change in DCI-IPG released per unit of insulin during OGTT (r = 0.47, P < 0.01). CONCLUSION: In women with PCOS, pioglitazone increased insulin-stimulated release of the DCI-IPG second messenger of insulin action, which may contribute to its insulin-sensitizing effect in these women.


Assuntos
Hipoglicemiantes/uso terapêutico , Fosfatos de Inositol/sangue , Insulina/uso terapêutico , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/tratamento farmacológico , Polissacarídeos/sangue , Tiazolidinedionas/uso terapêutico , Adolescente , Adulto , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Fosfatos de Inositol/deficiência , Insulina/administração & dosagem , Insulina/sangue , Antagonistas da Insulina/sangue , Resistência à Insulina , Pioglitazona , Polissacarídeos/deficiência , Tiazolidinedionas/administração & dosagem , Adulto Jovem
20.
Cell Signal ; 28(5): 384-390, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26852666

RESUMO

Resistance to insulin action is a key cause of diabetic complications, yet much remains unknown about the molecular mechanisms that contribute to the defect. Glucose-induced insulin resistance in peripheral tissues such as the retina is mediated in part by the hexosamine biosynthetic pathway (HBP). Glucosamine (GAM), a leading dietary supplement marketed to relieve the discomfort of osteoarthritis, is metabolized by the HBP, and in doing so bypasses the rate-limiting enzyme of the pathway. Thus, exogenous GAM consumption potentially exacerbates the resistance to insulin action observed with diabetes-induced hyperglycemia. In the present study, we evaluated the effect of GAM on insulin action in retinal Müller cells in culture. Addition of GAM to Müller cell culture repressed insulin-induced activation of the Akt/mTORC1 signaling pathway. However, the effect was not recapitulated by chemical inhibition to promote protein O-GlcNAcylation, nor was blockade of O-GlcNAcylation sufficient to prevent the effects of GAM. Instead, GAM induced ER stress and subsequent expression of the protein Regulated in DNA Damage and Development (REDD1), which was necessary for GAM to repress insulin-stimulated phosphorylation of Akt on Thr308. Overall, the findings support a model whereby GAM promotes ER stress in retinal Müller cells, resulting in elevated REDD1 expression and thus resistance to insulin action.


Assuntos
Células Ependimogliais/metabolismo , Glucosamina/farmacologia , Antagonistas da Insulina/farmacologia , Retina/metabolismo , Fatores de Transcrição/metabolismo , Acetilglucosamina/metabolismo , Animais , Células Cultivadas , Estresse do Retículo Endoplasmático , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/enzimologia , Insulina/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/citologia , Retina/enzimologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Treonina/metabolismo , Fatores de Transcrição/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA