Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(10)2022 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-35628359

RESUMO

Maintaining a robust, stable source of energy for doing chemical and physical work is essential to all living organisms. In eukaryotes, metabolic energy (ATP) production and consumption occurs in two separate compartments, the mitochondrial matrix and the cytosol. As a result, understanding eukaryotic metabolism requires knowledge of energy metabolism in each compartment and how metabolism in the two compartments is coordinated. Central to energy metabolism is the adenylate energy state ([ATP]/[ADP][Pi]). ATP is synthesized by oxidative phosphorylation (mitochondrial matrix) and glycolysis (cytosol) and each compartment provides the energy to do physical work and to drive energetically unfavorable chemical syntheses. The energy state in the cytoplasmic compartment has been established by analysis of near equilibrium metabolic reactions localized in that compartment. In the present paper, analysis is presented for energy-dependent reactions localized in the mitochondrial matrix using data obtained from both isolated mitochondria and intact tissues. It is concluded that the energy state ([ATP]f/[ADP]f[Pi]) in the mitochondrial matrix, calculated from the free (unbound) concentrations, is not different from the energy state in the cytoplasm. Corollaries are: (1) ADP in both the cytosol and matrix is selectively bound and the free concentrations are much lower than the total measured concentrations; and (2) under physiological conditions, the adenylate energy states in the mitochondrial matrix and cytoplasm are not substantially different.


Assuntos
Trifosfato de Adenosina , Eucariotos , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Citosol/metabolismo , Metabolismo Energético , Eucariotos/metabolismo
2.
Med Hypotheses ; 132: 109375, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31454640

RESUMO

Hyperbaric oxygen exposure is a recent hazzard for higher animals that originated as humans began underwater construction, exploration, and sports. Exposure can lead to abnormal brain EEG, convulsions, and death, the time to onset of each stage of pathology decreasing with increase in oxygen pressure. We provide evidence that hyperoxia, through oxidative phosphorylation, increases the energy state ([ATP]/[ADP][Pi]) of cells critical to providing glucose to cells behind the blood brain barrier (BBB). Brain cells without an absolute dependence on glucose metabolism; i.e. those having sufficient ATP synthesis using lactate and glutamate as oxidizable substrates, are not themselves very adversely affected by hyperoxia. The increased energy state and decrease in free [AMP], however, suppress glucose transport through the blood brain barrier (BBB) and into cells behind the BBB. Glucose has to pass in sequence through three steps of transport by facilitated diffusion and transporter activity for each step is regulated in part by AMP dependent protein kinase. The physiological role of this regulation is to increase glucose transport in response to hypoxia and/or systemic hypoglycemia. Hyperoxia, however, through unphysiological decrease in free [AMP] suppresses 1) glucose transport through the BBB (endothelial GLUT1 transporters) into cerebrospinal fluid (CSF); 2) glucose transport from CSF into cells behind the BBB (GLUT3 transporters) and (GLUT4 transporters). Cumulative suppression of glucose transport results in local regions of hypoglycemia and induces hypoglycemic failure. It is suggested that failure is initiated at axons and synapses with insufficient mitochondria to meet their energy requirements.


Assuntos
Encéfalo/patologia , Oxigenoterapia Hiperbárica/efeitos adversos , Hiperóxia/patologia , Hipoglicemia/etiologia , Trifosfato de Adenosina/metabolismo , Animais , Barreira Hematoencefálica , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 3/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Humanos , Hiperóxia/complicações , Camundongos , Mitocôndrias/metabolismo , Fosforilação
3.
Proc Natl Acad Sci U S A ; 116(37): 18684-18690, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31451647

RESUMO

Given the global epidemic in type 2 diabetes, novel antidiabetic drugs with increased efficacy and reduced side effects are urgently needed. Previous work has shown that M3 muscarinic acetylcholine (ACh) receptors (M3Rs) expressed by pancreatic ß cells play key roles in stimulating insulin secretion and maintaining physiological blood glucose levels. In the present study, we tested the hypothesis that a positive allosteric modulator (PAM) of M3R function can improve glucose homeostasis in mice by promoting insulin release. One major advantage of this approach is that allosteric agents respect the ACh-dependent spatiotemporal control of M3R activity. In this study, we first demonstrated that VU0119498, a drug known to act as a PAM at M3Rs, significantly augmented ACh-induced insulin release from cultured ß cells and mouse and human pancreatic islets. This stimulatory effect was absent in islets prepared from mice lacking M3Rs, indicative of the involvement of M3Rs. VU0119498 treatment of wild-type mice caused a significant increase in plasma insulin levels, accompanied by a striking improvement in glucose tolerance. These effects were mediated by ß-cell M3Rs, since they were absent in mutant mice selectively lacking M3Rs in ß cells. Moreover, acute VU0119498 treatment of obese, glucose-intolerant mice triggered enhanced insulin release and restored normal glucose tolerance. Interestingly, doses of VU0119498 that led to pronounced improvements in glucose homeostasis did not cause any significant side effects due to activation of M3Rs expressed by other peripheral cell types. Taken together, the data from this proof-of-concept study strongly suggest that M3R PAMs may become clinically useful as novel antidiabetic agents.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Agonistas Muscarínicos/farmacologia , Receptor Muscarínico M3/efeitos dos fármacos , Acetilcolina/metabolismo , Adulto , Regulação Alostérica/efeitos dos fármacos , Animais , Glicemia/análise , Glicemia/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/metabolismo , Humanos , Hipoglicemiantes/uso terapêutico , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Pessoa de Meia-Idade , Agonistas Muscarínicos/uso terapêutico , Obesidade/sangue , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Cultura Primária de Células , Estudo de Prova de Conceito , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Adulto Jovem
4.
Physiol Rep ; 5(12)2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28655753

RESUMO

A model for glucose sensing by pancreatic ß-cells is developed and compared with the available experimental data. The model brings together mathematical representations for the activities of the glucose sensor, glucokinase, and oxidative phosphorylation. Glucokinase produces glucose 6-phosphate (G-6-P) in an irreversible reaction that determines glycolytic flux. The primary products of glycolysis are NADH and pyruvate. The NADH is reoxidized and the reducing equivalents transferred to oxidative phosphorylation by the glycerol phosphate shuttle, and some of the pyruvate is oxidized by pyruvate dehydrogenase and enters the citric acid cycle. These reactions are irreversible and result in a glucose concentration-dependent reduction of the intramitochondrial NAD pool. This increases the electrochemical energy coupled to ATP synthesis and thereby the cellular energy state ([ATP]/[ADP][Pi]). ATP and Pi are 10-100 times greater than ADP, so the increase in energy state is primarily through decrease in ADP The decrease in ADP is considered responsible for altering ion channel conductance and releasing insulin. Applied to the reported glucose concentration-dependent release of insulin by perifused islet preparations (Doliba et al. 2012), the model predicts that the dependence of insulin release on ADP is strongly cooperative with a threshold of about 30 µmol/L and a negative Hill coefficient near -5.5. The predicted cellular energy state, ADP, creatine phosphate/creatine ratio, and cytochrome c reduction, including their dependence on glucose concentration, are consistent with experimental data. The ability of the model to predict behavior consistent with experiment is an invaluable resource for understanding glucose sensing and planning experiments.


Assuntos
Trifosfato de Adenosina/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Modelos Biológicos , Termodinâmica , Animais , Humanos , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Fosforilação Oxidativa
5.
Diabetes ; 66(7): 1901-1913, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28442472

RESUMO

Loss-of-function mutations of ß-cell KATP channels cause the most severe form of congenital hyperinsulinism (KATPHI). KATPHI is characterized by fasting and protein-induced hypoglycemia that is unresponsive to medical therapy. For a better understanding of the pathophysiology of KATPHI, we examined cytosolic calcium ([Ca2+] i ), insulin secretion, oxygen consumption, and [U-13C]glucose metabolism in islets isolated from the pancreases of children with KATPHI who required pancreatectomy. Basal [Ca2+] i and insulin secretion were higher in KATPHI islets compared with controls. Unlike controls, insulin secretion in KATPHI islets increased in response to amino acids but not to glucose. KATPHI islets have an increased basal rate of oxygen consumption and mitochondrial mass. [U-13C]glucose metabolism showed a twofold increase in alanine levels and sixfold increase in 13C enrichment of alanine in KATPHI islets, suggesting increased rates of glycolysis. KATPHI islets also exhibited increased serine/glycine and glutamine biosynthesis. In contrast, KATPHI islets had low γ-aminobutyric acid (GABA) levels and lacked 13C incorporation into GABA in response to glucose stimulation. The expression of key genes involved in these metabolic pathways was significantly different in KATPHI ß-cells compared with control, providing a mechanism for the observed changes. These findings demonstrate that the pathophysiology of KATPHI is complex, and they provide a framework for the identification of new potential therapeutic targets for this devastating condition.


Assuntos
Cálcio/metabolismo , Hiperinsulinismo Congênito/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Consumo de Oxigênio , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Sulfonilureias/metabolismo , Alanina/metabolismo , Isótopos de Carbono , Estudos de Casos e Controles , Hiperinsulinismo Congênito/genética , Hiperinsulinismo Congênito/cirurgia , Feminino , Citometria de Fluxo , Expressão Gênica , Glutamina/biossíntese , Glicina/biossíntese , Glicólise/genética , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Secreção de Insulina , Células Secretoras de Insulina/ultraestrutura , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Canais KATP/genética , Canais KATP/metabolismo , Masculino , Metabolômica , Microscopia Eletrônica de Transmissão , Mutação , Pancreatectomia , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Serina/biossíntese , Receptores de Sulfonilureias/genética , Ácido gama-Aminobutírico/metabolismo
6.
J Fluoresc ; 27(5): 1621-1631, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28432632

RESUMO

Five variants of glucokinase (ATP-D-hexose-6-phosphotransferase, EC 2.7.1.1) including wild type and single Trp mutants with the Trp residue at positions 65, 99, 167 and 257 were prepared. The fluorescence of Trp in all locations studied showed intensity changes when glucose bound, indicating that conformational change occurs globally over the entire protein. While the fluorescence quantum yield changes upon glucose binding, the enzyme's absorption spectra, emission spectra and fluorescence lifetimes change very little. These results are consistent with the existence of a dark complex for excited state Trp. Addition of glycerol, L-glucose, sucrose, or trehalose increases the binding affinity of glucose to the enzyme and increases fluorescence intensity. The effect of these osmolytes is thought to shift the protein conformation to a condensed, high affinity form. Based upon these results, we consider the nature of quenching of the Trp excited state. Amide groups are known to quench indole fluorescence and amides of the polypeptide chain make interact with excited state Trp in the relatively unstructured, glucose-free enzyme. Also, removal of water around the aromatic ring by addition of glucose substrate or osmolyte may reduce the quenching.


Assuntos
Fluorescência , Glucoquinase/química , Conformação Proteica , Triptofano/química , Glucoquinase/genética , Glucoquinase/metabolismo , Humanos , Mutação , Espectrometria de Fluorescência , Especificidade por Substrato , Triptofano/metabolismo
7.
Nat Commun ; 8: 14295, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28145434

RESUMO

ß-arrestins are critical signalling molecules that regulate many fundamental physiological functions including the maintenance of euglycemia and peripheral insulin sensitivity. Here we show that inactivation of the ß-arrestin-2 gene, barr2, in ß-cells of adult mice greatly impairs insulin release and glucose tolerance in mice fed with a calorie-rich diet. Both glucose and KCl-induced insulin secretion and calcium responses were profoundly reduced in ß-arrestin-2 (barr2) deficient ß-cells. In human ß-cells, barr2 knockdown abolished glucose-induced insulin secretion. We also show that the presence of barr2 is essential for proper CAMKII function in ß-cells. Importantly, overexpression of barr2 in ß-cells greatly ameliorates the metabolic deficits displayed by mice consuming a high-fat diet. Thus, our data identify barr2 as an important regulator of ß-cell function, which may serve as a new target to improve ß-cell function.


Assuntos
Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais/genética , beta-Arrestina 2/genética , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Dieta Hiperlipídica , Expressão Gênica , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , beta-Arrestina 2/metabolismo
8.
Am J Physiol Endocrinol Metab ; 302(1): E87-E102, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21952036

RESUMO

It was reported previously that isolated human islets from individuals with type 2 diabetes mellitus (T2DM) show reduced glucose-stimulated insulin release. To assess the possibility that impaired bioenergetics may contribute to this defect, glucose-stimulated respiration (Vo(2)), glucose usage and oxidation, intracellular Ca(2+), and insulin secretion (IS) were measured in pancreatic islets isolated from three healthy and three type 2 diabetic organ donors. Isolated mouse and rat islets were studied for comparison. Islets were exposed to a "staircase" glucose stimulus, whereas IR and Vo(2) were measured. Vo(2) of human islets from normals and diabetics increased sigmoidally from equal baselines of 0.25 nmol/100 islets/min as a function of glucose concentration. Maximal Vo(2) of normal islets at 24 mM glucose was 0.40 ± 0.02 nmol·min(-1)·100 islets(-1), and the glucose S(0.5) was 4.39 ± 0.10 mM. The glucose stimulation of respiration of islets from diabetics was lower, V(max) of 0.32 ± 0.01 nmol·min(-1)·100 islets(-1), and the S(0.5) shifted to 5.43 ± 0.13 mM. Glucose-stimulated IS and the rise of intracellular Ca(2+) were also reduced in diabetic islets. A clinically effective glucokinase activator normalized the defective Vo(2), IR, and free calcium responses during glucose stimulation in islets from type 2 diabetics. The body of data shows that there is a clear relationship between the pancreatic islet energy (ATP) production rate and IS. This relationship was similar for normal human, mouse, and rat islets and the data for all species fitted a single sigmoidal curve. The shared threshold rate for IS was ∼13 pmol·min(-1)·islet(-1). Exendin-4, a GLP-1 analog, shifted the ATP production-IS curve to the left and greatly potentiated IS with an ATP production rate threshold of ∼10 pmol·min(-1)·islet(-1). Our data suggest that impaired ß-cell bioenergetics resulting in greatly reduced ATP production is critical in the molecular pathogenesis of type 2 diabetes mellitus.


Assuntos
Benzenoacetamidas/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Ativadores de Enzimas/farmacologia , Glucoquinase/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Adulto , Animais , Sinalização do Cálcio/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Exenatida , Feminino , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Glucoquinase/química , Glicólise/efeitos dos fármacos , Humanos , Hipoglicemiantes/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Fosforilação Oxidativa/efeitos dos fármacos , Peptídeos/farmacologia , Ratos , Especificidade da Espécie , Técnicas de Cultura de Tecidos , Peçonhas/farmacologia
9.
Diabetes ; 60(12): 3175-85, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22028181

RESUMO

OBJECTIVE: The posttranslational regulation of glucokinase (GK) differs in hepatocytes and pancreatic ß-cells. We tested the hypothesis that GK mutants that cause maturity-onset diabetes of the young (GK-MODY) show compromised activity and posttranslational regulation in ß-cells. RESEARCH DESIGN AND METHODS: Activity and protein expression of GK-MODY and persistent hyperinsulinemic hypoglycemia of infancy (PHHI) mutants were studied in ß-cell (MIN6) and non-ß-cell (H4IIE) models. Binding of GK to phosphofructo-2-kinase, fructose-2,6-bisphosphatase (PFK2/FBPase2) was studied by bimolecular fluorescence complementation in cell-based models. RESULTS: Nine of 11 GK-MODY mutants that have minimal effect on enzyme kinetics in vitro showed decreased specific activity relative to wild type when expressed in ß-cells. A subset of these were stable in non-ß-cells but showed increased inactivation in conditions of oxidative stress and partial reversal of inactivation by dithiothreitol. Unlike the GK-MODY mutants, four of five GK-PHHI mutants had similar specific activity to wild type and Y214C had higher activity than wild type. The GK-binding protein PFK2/FBPase2 protected wild-type GK from oxidative inactivation and the decreased stability of GK-MODY mutants correlated with decreased interaction with PFK2/FBPase2. CONCLUSIONS: Several GK-MODY mutants show posttranslational defects in ß-cells characterized by increased susceptibility to oxidative stress and/or protein instability. Regulation of GK activity through modulation of thiol status may be a physiological regulatory mechanism for the control of GK activity in ß-cells.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucoquinase/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Glucoquinase/genética , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Mutação , Óxido Nítrico/metabolismo , Estresse Oxidativo , Fosfofrutoquinase-2/metabolismo , Ligação Proteica , Ratos , Ratos Wistar
10.
J Biol Chem ; 286(39): 34164-74, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21813650

RESUMO

Glutamate dehydrogenase (GDH) catalyzes the oxidative deamination of L-glutamate and, in animals, is extensively regulated by a number of metabolites. Gain of function mutations in GDH that abrogate GTP inhibition cause the hyperinsulinism/hyperammonemia syndrome (HHS), resulting in increased pancreatic ß-cell responsiveness to leucine and susceptibility to hypoglycemia following high protein meals. We have previously shown that two of the polyphenols from green tea (epigallocatechin gallate (EGCG) and epicatechin gallate (ECG)) inhibit GDH in vitro and that EGCG blocks GDH-mediated insulin secretion in wild type rat islets. Using structural and site-directed mutagenesis studies, we demonstrate that ECG binds to the same site as the allosteric regulator, ADP. Perifusion assays using pancreatic islets from transgenic mice expressing a human HHS form of GDH demonstrate that the hyperresponse to glutamine caused by dysregulated GDH is blocked by the addition of EGCG. As observed in HHS patients, these transgenic mice are hypersensitive to amino acid feeding, and this is abrogated by oral administration of EGCG prior to challenge. Finally, the low basal blood glucose level in the HHS mouse model is improved upon chronic administration of EGCG. These results suggest that this common natural product or some derivative thereof may prove useful in controlling this genetic disorder. Of broader clinical implication is that other groups have shown that restriction of glutamine catabolism via these GDH inhibitors can be useful in treating various tumors. This HHS transgenic mouse model offers a highly useful means to test these agents in vivo.


Assuntos
Difosfato de Adenosina/metabolismo , Anticarcinógenos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Catequina/análogos & derivados , Glutamato Desidrogenase/metabolismo , Ilhotas Pancreáticas/metabolismo , Chá/química , Difosfato de Adenosina/genética , Animais , Anticarcinógenos/química , Antineoplásicos Fitogênicos/química , Domínio Catalítico/genética , Catequina/química , Catequina/farmacologia , Glucose/genética , Glucose/metabolismo , Glutamato Desidrogenase/genética , Glutamina/genética , Glutamina/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Ratos
11.
Proc Natl Acad Sci U S A ; 108(29): 12030-5, 2011 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-21730150

RESUMO

Death of pancreatic ß cells is a pathological hallmark of type 1 diabetes (T1D). However, the molecular mechanisms of ß cell death and its regulation are poorly understood. Here we describe a unique regulatory pathway of ß cell death that comprises microRNA-21, its target tumor suppressor PDCD4, and its upstream transcriptional activator nuclear factor-κB (NF-κB). In pancreatic ß cells, c-Rel and p65 of the NF-κB family activated the mir21 gene promoter and increased miR-21 RNA levels; miR-21 in turn decreased the level of PDCD4, which is able to induce cell death through the Bax family of apoptotic proteins. Consequently, PDCD4 deficiency in pancreatic ß cells renders them resistant to death, and PDCD4 deficiency in NOD or C57BL/6 mice conferred resistance to spontaneous diabetes and diabetes induced by autoimmune T cells or the ß cell toxin streptozotocin (STZ). Thus, the NF-κB-microRNA-21-PDCD4 axis plays a crucial role in T1D and represents a unique therapeutic target for treating the disease.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Morte Celular/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Regulação da Expressão Gênica/imunologia , Células Secretoras de Insulina/fisiologia , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Proteínas de Ligação a RNA/metabolismo , Análise de Variância , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/imunologia , Morte Celular/genética , Primers do DNA/genética , Diabetes Mellitus Tipo 1/prevenção & controle , Citometria de Fluxo , Immunoblotting , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , MicroRNAs/imunologia , NF-kappa B/imunologia , Proteínas de Ligação a RNA/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Am J Physiol Endocrinol Metab ; 299(3): E475-85, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20606076

RESUMO

Fatty acids, acetylcholine, and GLP-1 enhance insulin secretion in a glucose-dependent manner. However, the interplay between glucose, fatty acids, and the neuroendocrine regulators of insulin secretion is not well understood. Therefore, we studied the acute effects of PA (alone or in combination with glucose, acetylcholine, or GLP-1) on isolated cultured mouse islets. Two different sets of experiments were designed. In one, a fixed concentration of 0.5 mM of PA bound to 0.15 mM BSA was used; in the other, a PA ramp from 0 to 0.5 mM was applied at a fixed albumin concentration of 0.15 mM so that the molar PA/BSA ratio changed within the physiological range. At a fixed concentration of 0.5 mM, PA markedly inhibited acetylcholine-stimulated insulin release, the rise of intracellular Ca(2+), and enhancement of cAMP production but did not influence the effects of GLP-1 on these parameters of islet cell function. 2-ADB, an IP(3) receptor inhibitor, reduced the effect of acetylcholine on insulin secretion and reversed the effect of PA on acetylcholine-stimulated insulin release. Islet perfusion for 35-40 min with 0.5 mM PA significantly reduced the calcium storage capacity of ER measured by the thapsigargin-induced Ca(2+) release. Oxygen consumption due to low but not high glucose was reduced by PA. When a PA ramp from 0 to 0.5 mM was applied in the presence of 8 mM glucose, PA at concentrations as low as 50 microM significantly augmented glucose-stimulated insulin release and markedly reduced acetylcholine's effects on hormone secretion. We thus demonstrate that PA acutely reduces the total oxygen consumption response to glucose, glucose-dependent acetylcholine stimulation of insulin release, Ca(2+), and cAMP metabolism, whereas GLP-1's actions on these parameters remain unaffected or potentiated. We speculate that acute emptying of the ER calcium by PA results in decreased glucose stimulation of respiration and acetylcholine potentiation of insulin secretion.


Assuntos
Acetilcolina/antagonistas & inibidores , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Ácido Palmítico/farmacologia , Acetilcolina/metabolismo , Animais , Cálcio/metabolismo , AMP Cíclico/metabolismo , Técnicas In Vitro , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Consumo de Oxigênio/fisiologia
13.
J Biol Chem ; 284(18): 12246-57, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19265200

RESUMO

Pancreatic islet beta-cell dysfunction is a signature feature of Type 2 diabetes pathogenesis. Consequently, knowledge of signals that regulate beta-cell function is of immense clinical relevance. Transforming growth factor (TGF)-beta signaling plays a critical role in pancreatic development although the role of this pathway in the adult pancreas is obscure. Here, we define an important role of the TGF-beta pathway in regulation of insulin gene transcription and beta-cell function. We identify insulin as a TGF-beta target gene and show that the TGF-beta signaling effector Smad3 occupies the insulin gene promoter and represses insulin gene transcription. In contrast, Smad3 small interfering RNAs relieve insulin transcriptional repression and enhance insulin levels. Transduction of adenoviral Smad3 into primary human and non-human primate islets suppresses insulin content, whereas, dominant-negative Smad3 enhances insulin levels. Consistent with this, Smad3-deficient mice exhibit moderate hyperinsulinemia and mild hypoglycemia. Moreover, Smad3 deficiency results in improved glucose tolerance and enhanced glucose-stimulated insulin secretion in vivo. In ex vivo perifusion assays, Smad3-deficient islets exhibit improved glucose-stimulated insulin release. Interestingly, Smad3-deficient islets harbor an activated insulin-receptor signaling pathway and TGF-beta signaling regulates expression of genes involved in beta-cell function. Together, these studies emphasize TGF-beta/Smad3 signaling as an important regulator of insulin gene transcription and beta-cell function and suggest that components of the TGF-beta signaling pathway may be dysregulated in diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Transdução de Sinais , Proteína Smad3/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucose/genética , Glucose/metabolismo , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hipoglicemia/genética , Hipoglicemia/metabolismo , Insulina/genética , Camundongos , Camundongos Knockout , Primatas , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética
14.
J Biol Chem ; 283(38): 25786-93, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18635551

RESUMO

Congenital hyperinsulinism is a disorder of pancreatic beta-cell function characterized by failure to suppress insulin secretion in the setting of hypoglycemia, resulting in brain damage or death if untreated. Loss-of-function mutations in the K(ATP) channel (composed of two subunits: Kir6.2 and SUR-1) are responsible for the most common and severe form of congenital hyperinsulinism. Most patients are unresponsive to available medical therapy and require palliative pancreatectomy. Similar to the human condition, the SUR-1(-/-) mouse is hypoglycemic when fasted and hyperglycemic when glucose-loaded. We have previously reported that the glucagon-like peptide-1 receptor antagonist exendin-(9-39) raises fasting blood glucose in normal mice. Here we examine the effect of exendin-(9-39) on fasting blood glucose in SUR-1(-/-) mice. Mice were randomized to receive exendin-(9-39) or vehicle. Fasting blood glucose levels in SUR-1(-/-) mice treated with exendin-(9-39) were significantly higher than in vehicle-treated mice and not different from wild-type littermates. Exendin-(9-39) did not further worsen glucose tolerance and had no effect on body weight and insulin sensitivity. Isolated islet perifusion studies demonstrated that exendin-(9-39) blocked amino acid-stimulated insulin secretion, which is abnormally increased in SUR-1(-/-) islets. Furthermore, cAMP content in SUR-1(-/-) islets was reduced by exendin-(9-39) both basally and when stimulated by amino acids, whereas cytosolic calcium levels were not affected. These findings suggest that cAMP plays a key role in K(ATP)-independent insulin secretion and that the GLP-1 receptor is constitutively active in SUR-1(-/-) beta-cells. Our findings indicate that exendin-(9-39) normalizes fasting hypoglycemia in SUR-1(-/-) mice via a direct effect on insulin secretion, thereby raising exendin-(9-39) as a potential therapeutic agent for K(ATP) hyperinsulinism.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Regulação da Expressão Gênica , Hipoglicemia/metabolismo , Células Secretoras de Insulina/citologia , Insulina/metabolismo , Fragmentos de Peptídeos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Receptores de Droga/genética , Receptores de Glucagon/metabolismo , Animais , AMP Cíclico/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/metabolismo , Hipoglicemia/etiologia , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Fragmentos de Peptídeos/química , Receptores de Sulfonilureias
15.
J Biol Chem ; 283(25): 17238-49, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18445600

RESUMO

Pancreatic beta cells are hyper-responsive to amino acids but have decreased glucose sensitivity after deletion of the sulfonylurea receptor 1 (SUR1) both in man and mouse. It was hypothesized that these defects are the consequence of impaired integration of amino acid, glucose, and energy metabolism in beta cells. We used gas chromatography-mass spectrometry methodology to study intermediary metabolism of SUR1 knock-out (SUR1(-/-)) and control mouse islets with d-[U-(13)C]glucose as substrate and related the results to insulin secretion. The levels and isotope labeling of alanine, aspartate, glutamate, glutamine, and gamma-aminobutyric acid (GABA) served as indicators of intermediary metabolism. We found that the GABA shunt of SUR1(-/-) islets is blocked by about 75% and showed that this defect is due to decreased glutamate decarboxylase synthesis, probably caused by elevated free intracellular calcium. Glutaminolysis stimulated by the leucine analogue d,l-beta-2-amino-2-norbornane-carboxylic acid was, however, enhanced in SUR1(-/-) and glyburide-treated SUR1(+/+) islets. Glucose oxidation and pyruvate cycling was increased in SUR1(-/-) islets at low glucose but was the same as in controls at high glucose. Malic enzyme isoforms 1, 2, and 3, involved in pyruvate cycling, were all expressed in islets. High glucose lowered aspartate and stimulated glutamine synthesis similarly in controls and SUR1(-/-) islets. The data suggest that the interruption of the GABA shunt and the lack of glucose regulation of pyruvate cycling may cause the glucose insensitivity of the SUR1(-/-) islets but that enhanced basal pyruvate cycling, lowered GABA shunt flux, and enhanced glutaminolytic capacity may sensitize the beta cells to amino acid stimulation.


Assuntos
Trifosfato de Adenosina/química , Glucose/metabolismo , Glutamina/química , Potássio/química , Piruvatos/química , Ácido gama-Aminobutírico/metabolismo , Aminoácidos/química , Animais , Cromatografia Gasosa-Espectrometria de Massas/métodos , Genótipo , Glutamato Descarboxilase/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Oxigênio/metabolismo
16.
J Histochem Cytochem ; 55(6): 555-66, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17283370

RESUMO

A recent report provides new evidence for the presence of glucokinase (GK) in the anterior pituitary. In the present study, immunohistochemistry was used to identify the cells containing GK in the pituitary of rats and monkeys. In rats, GK was detected as a generalized cytoplasmic staining in a discrete population of cells in the anterior pituitary. In colocalization experiments, the majority of cells expressing follicle-stimulating hormone (FSH) or luteinizing hormone (LH) also contained GK. In addition to the gonadotropes, GK was observed in a subpopulation of corticotropes and thyrotropes. GK was not detected in cells expressing growth hormone or prolactin. In monkeys, GK was also observed in a discrete population of cells. Intracellular distribution differed from the rat in that GK in most cells was concentrated in a perinuclear location that appeared to be associated with the Golgi apparatus. However, similar to rats, colocalization experiments showed that the majority of cells expressing FSH or LH also contained GK. In addition to the gonadotropes, GK was observed in a subpopulation of corticotropes and thyrotropes. In the monkey, only a few cells had generalized cytoplasmic staining for GK. These experiments provide further evidence for the presence of GK in the anterior pituitary. Although some corticotropes and thyrotropes contained GK, the predominant cell type expressing GK was gonadotropes. In view of the generally accepted role of GK as a glucose sensor in a variety of cells including the insulin-producing pancreatic beta-cells as the prototypical example, it is hypothesized that hormone synthesis and/or release in pituitary cells containing GK may be directly influenced by blood glucose.


Assuntos
Glucoquinase/metabolismo , Imuno-Histoquímica/métodos , Adeno-Hipófise/enzimologia , Animais , Feminino , Hormônio Foliculoestimulante/metabolismo , Gonadotropinas Hipofisárias/metabolismo , Hormônio Luteinizante/metabolismo , Macaca fascicularis , Masculino , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Tireotropina/metabolismo
17.
Am J Physiol Endocrinol Metab ; 292(6): E1507-19, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17264232

RESUMO

Fuel stimulation of insulin secretion from pancreatic beta-cells is thought to be mediated by metabolic coupling factors that are generated by energized mitochondria, including protons, adenine nucleotides, and perhaps certain amino acids (AA), as for instance aspartate, glutamate, or glutamine (Q). The goal of the present study was to evaluate the role of such factors when insulin release (IR) is stimulated by glucose or AA, alone or combined, using (31)P, (23)Na and (1)H NMR technology, respirometry, and biochemical analysis to study the metabolic events that occur in continuously superfused mouse beta-HC9 cells contained in agarose beads and enhanced by the phosphodiesterase inhibitor IBMX. Exposing beta-HC9 cells to high glucose or 3.5 mM of a physiological mixture of 18 AA (AAM) plus 2 mM glutamine caused a marked stimulation of insulin secretion associated with increased oxygen consumption, cAMP release, and phosphorylation potential as evidenced by higher phosphocreatine and lower P(i) peak areas of (31)P NMR spectra. Diazoxide blocked stimulation of IR completely, suggesting involvement of ATP-dependent potassium (K(ATP)) channels in this process. However, levels of MgATP and MgADP concentrations, which regulate channel activity, changed only slowly and little, whereas the rate of insulin release increased fast and very markedly. The involvement of other candidate coupling factors was therefore considered. High glucose or AAM + Q increased pH(i). The availability of temporal pH profiles allowed the precise computation of the phosphate potential (ATP/P(i) x ADP) in fuel-stimulated IR. Intracellular Na+ levels were greatly elevated by AAM + Q. However, glutamine alone or together with 2-amino-2-norbornanecarboxylic acid (which activates glutamate dehydrogenase) decreased beta-cell Na levels. Stimulation of beta-cells by glucose in the presence of AAM + Q (0.5 mM) was associated with rising cellular concentrations of glutamate and glutamine and strikingly lower aspartate levels. Methionine sulfoximine, an inhibitor of glutamine synthetase, blocked the glucose enhancement of AMM + Q-induced IR and associated changes in glutamine and aspartate but did not prevent the accumulation of glutamate. The results of this study demonstrate again that an increased phosphate potential and a functional K(ATP) channel are essential for metabolic coupling during fuel-stimulated insulin release but illustrate that determining the identity and relative importance of all participating coupling factors and second messengers remains a challenge largely unmet.


Assuntos
Aminoácidos/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Aminoácidos/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Citosol/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Glucose/administração & dosagem , Glucose/farmacologia , Glutamina/farmacologia , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Camundongos , Consumo de Oxigênio/efeitos dos fármacos , Fosfatos/metabolismo , Fosforilação , Canais de Potássio/metabolismo , Sódio/metabolismo , Fatores de Tempo
18.
Diabetes ; 55(7): 1923-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16804059

RESUMO

Enzymatic activity of glucokinase was demonstrated, quantitated, and characterized kinetically in rat and mouse pituitary extracts using a highly specific and sensitive spectrometric assay. A previously proposed hypothesis that the glucokinase gene might be expressed in the pituitary corticotrophic cells was therefore reexamined using mRNA in situ hybridization and immunohistochemical techniques. No evidence was found that corticotrophs are glucokinase positive, and the identity of glucokinase-expressing cells remains to be determined. The findings do, however, suggest a novel hypothesis that a critical subgroup of anterior pituitary cells might function as glucose sensor cells and that direct fuel regulation of such cells may modify the classical indirect neuroendocrine pathways that are known to control hormone secretion from anterior pituitary cells.


Assuntos
Glucoquinase/genética , Adeno-Hipófise/enzimologia , Hormônio Adrenocorticotrópico/genética , Animais , Técnicas Biossensoriais , Feminino , Regulação Enzimológica da Expressão Gênica , Hormônio do Crescimento/genética , Hibridização In Situ , Cinética , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Pró-Opiomelanocortina/genética , RNA Mensageiro/genética , Ratos
19.
J Biol Chem ; 281(22): 15064-72, 2006 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-16574664

RESUMO

Glutamate dehydrogenase (GDH) plays an important role in insulin secretion as evidenced in children by gain of function mutations of this enzyme that cause a hyperinsulinism-hyperammonemia syndrome (GDH-HI) and sensitize beta-cells to leucine stimulation. GDH transgenic mice were generated to express the human GDH-HI H454Y mutation and human wild-type GDH in islets driven by the rat insulin promoter. H454Y transgene expression was confirmed by increased GDH enzyme activity in islets and decreased sensitivity to GTP inhibition. The H454Y GDH transgenic mice had hypoglycemia with normal growth rates. H454Y GDH transgenic islets were more sensitive to leucine- and glutamine-stimulated insulin secretion but had decreased response to glucose stimulation. The fluxes via GDH and glutaminase were measured by tracing 15N flux from [2-15N]glutamine. The H454Y transgene in islets had higher insulin secretion in response to glutamine alone and had 2-fold greater GDH flux. High glucose inhibited both glutaminase and GDH flux, and leucine could not override this inhibition. 15NH4Cl tracing studies showed 15N was not incorporated into glutamate in either H454Y transgenic or normal islets. In conclusion, we generated a GDH-HI disease mouse model that has a hypoglycemia phenotype and confirmed that the mutation of H454Y is disease causing. Stimulation of insulin release by the H454Y GDH mutation or by leucine activation is associated with increased oxidative deamination of glutamate via GDH. This study suggests that GDH functions predominantly in the direction of glutamate oxidation rather than glutamate synthesis in mouse islets and that this flux is tightly controlled by glucose.


Assuntos
Glutamato Desidrogenase/genética , Insulina/metabolismo , Mutação , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Glucose/farmacologia , Glutamato Desidrogenase/antagonistas & inibidores , Glutamato Desidrogenase/metabolismo , Glutamina/farmacologia , Guanosina Trifosfato/farmacologia , Humanos , Hiperinsulinismo/enzimologia , Hiperinsulinismo/genética , Hiperinsulinismo/fisiopatologia , Técnicas In Vitro , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Cinética , Leucina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
J Biol Chem ; 281(15): 10214-21, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16476731

RESUMO

Insulin secretion by pancreatic beta-cells is stimulated by glucose, amino acids, and other metabolic fuels. Glutamate dehydrogenase (GDH) has been shown to play a regulatory role in this process. The importance of GDH was underscored by features of hyperinsulinemia/hyperammonemia syndrome, where a dominant mutation causes the loss of inhibition by GTP and ATP. Here we report the effects of green tea polyphenols on GDH and insulin secretion. Of the four compounds tested, epigallocatechin gallate (EGCG) and epicatechin gallate were found to inhibit GDH with nanomolar ED(50) values and were therefore found to be as potent as the physiologically important inhibitor GTP. Furthermore, we have demonstrated that EGCG inhibits BCH-stimulated insulin secretion, a process that is mediated by GDH, under conditions where GDH is no longer inhibited by high energy metabolites. EGCG does not affect glucose-stimulated insulin secretion under high energy conditions where GDH is probably fully inhibited. We have further shown that these compounds act in an allosteric manner independent of their antioxidant activity and that the beta-cell stimulatory effects are directly correlated with glutamine oxidation. These results demonstrate that EGCG, much like the activator of GDH (BCH), can facilitate dissecting the complex regulation of insulin secretion by pharmacologically modulating the effects of GDH.


Assuntos
Inibidores Enzimáticos/farmacologia , Flavonoides/química , Glutamato Desidrogenase/antagonistas & inibidores , Insulina/metabolismo , Fenóis/química , Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Animais , Bovinos , Relação Dose-Resposta a Droga , Glutamato Desidrogenase/metabolismo , Guanosina Trifosfato/química , Hiperamonemia/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Cinética , Leucina/química , Masculino , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Consumo de Oxigênio , Perfusão , Polifenóis , Conformação Proteica , Ratos , Ratos Wistar , Chá , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA