Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem J ; 459(3): 551-64, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24568320

RESUMO

Hepatic glucose phosphorylation by GK (glucokinase) is regulated by GKRP (GK regulatory protein). GKRP forms a cytosolic complex with GK followed by nuclear import and storage, leading to inhibition of GK activity. This process is initiated by low glucose, but reversed nutritionally by high glucose and fructose or pharmacologically by GKAs (GK activators) and GKRPIs (GKRP inhibitors). To study the regulation of this process by glucose, fructose-phosphate esters and a GKA, we measured the TF (tryptophan fluorescence) of human WT (wild-type) and GKRP-P446L (a mutation associated with high serum triacylglycerol) in the presence of non-fluorescent GK with its tryptophan residues mutated. Titration of GKRP-WT by GK resulted in a sigmoidal increase in TF, suggesting co-operative PPIs (protein-protein interactions) perhaps due to the hysteretic nature of GK. The affinity of GK for GKRP was decreased and binding co-operativity increased by glucose, fructose 1-phosphate and GKA, reflecting disruption of the GK-GKRP complex. Similar studies with GKRP-P446L showed significantly different results compared with GKRP-WT, suggesting impairment of complex formation and nuclear storage. The results of the present TF-based biophysical analysis of PPIs between GK and GKRP suggest that hepatic glucose metabolism is regulated by a metabolite-sensitive drug-responsive co-operative molecular switch, involving complex formation between these two allosterically regulated proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glucoquinase/metabolismo , Modelos Moleculares , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Regulação Alostérica , Substituição de Aminoácidos , Frutosefosfatos/metabolismo , Glucoquinase/química , Glucoquinase/genética , Glucose/metabolismo , Humanos , Ligantes , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Redobramento de Proteína , Estabilidade Proteica , Transporte Proteico , Desdobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometria de Fluorescência , Triptofano/química
2.
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
3.
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
4.
Am J Physiol Endocrinol Metab ; 291(3): E525-35, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16638820

RESUMO

Neural and endocrine factors (i.e., Ach and GLP-1) restore defective glucose-stimulated insulin release in pancreatic islets lacking sulfonylurea type 1 receptors (SUR1(-/-)) (Doliba NM, Qin W, Vatamaniuk MZ, Li C, Zelent D, Najafi H, Buettger CW, Collins HW, Carr RD, Magnuson MA, and Matschinsky FM. Am J Physiol Endocrinol Metab 286: E834-E843, 2004). The goal of the present study was to assess fuel-induced respiration in SUR1(-/-) islets and to correlate it with changes in intracellular Ca(2+), insulin, and glucagon secretion. By use of a method based on O(2) quenching of phosphorescence, the O(2) consumption rate (OCR) of isolated islets was measured online in a perifusion system. Basal insulin release (IR) was 7-10 times higher in SUR1(-/-) compared with control (CON) islets, but the OCR was comparable. The effect of high glucose (16.7 mM) on IR and OCR was markedly reduced in SUR1(-/-) islets compared with CON. Ach (0.5 microM) in the presence of 16.7 mM glucose caused a large burst of IR in CON and SUR1(-/-) islets with minor changes in OCR in both groups of islets. In SUR1(-/-) islets, high glucose failed to inhibit glucagon secretion during stimulation with amino acids or Ach. We conclude that 1) reduced glucose responsiveness of SUR1(-/-) islets may be in part due to impaired energetics, as evidenced by significant decrease in glucose-stimulated OCR; 2) elevated intracellular Ca(2+) levels may contribute to altered insulin and glucagon secretion in SUR1(-/-) islets; and 3) The amplitudes of the changes in OCR during glucose and Ach stimulation do not correlate with IR in normal and SUR1(-/-) islets suggesting that the energy requirements for exocytosis are minor compared with other ATP-consuming reactions.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Respiração Celular/fisiologia , Fibras Colinérgicas/fisiologia , Glucose/farmacologia , Hormônios/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Acetilcolina/farmacologia , Acetilcolinesterase/genética , Aminoácidos/farmacologia , Animais , Cálcio/metabolismo , Respiração Celular/efeitos dos fármacos , Expressão Gênica/genética , Glucagon/metabolismo , Transportador de Glucose Tipo 2/genética , Glibureto/farmacologia , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Canal de Potássio Kv1.3 , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Consumo de Oxigênio/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Droga , Receptores de Sulfonilureias
5.
Am J Physiol Endocrinol Metab ; 286(5): E834-43, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14736703

RESUMO

Inhibition of ATP-sensitive K+ (K(ATP)) channels by an increase in the ATP/ADP ratio and the resultant membrane depolarization are considered essential in the process leading to insulin release (IR) from pancreatic beta-cells stimulated by glucose. It is therefore surprising that mice lacking the sulfonylurea type 1 receptor (SUR1-/-) in beta-cells remain euglycemic even though the knockout is expected to cause hypoglycemia. To complicate matters, isolated islets of SUR1-/- mice secrete little insulin in response to high glucose, which extrapolates to hyperglycemia in the intact animal. It remains thus unexplained how euglycemia is maintained. In recognition of the essential role of neural and endocrine regulation of IR, we evaluated the effects of acetylcholine (ACh) and glucagon-like peptide-1 (GLP-1) on IR and free intracellular Ca2+ concentration ([Ca2+]i) of freshly isolated or cultured islets of SUR1-/- mice and B6D2F1 controls (SUR1+/+). IBMX, a phosphodiesterase inhibitor, was also used to explore cAMP-dependent signaling in IR. Most striking, and in contrast to controls, SUR1-/-) islets are hypersensitive to ACh and IBMX, as demonstrated by a marked increase of IR even in the absence of glucose. The hypersensitivity to ACh was reproduced in control islets by depolarization with the SUR1 inhibitor glyburide. Pretreatment of perifused SUR1-/- islets with ACh or IBMX restored glucose stimulation of IR, an effect expectedly insensitive to diazoxide. The calcium channel blocker verapamil reduced but did not abolish ACh-stimulated IR, supporting a role for intracellular Ca2+ stores in stimulus-secretion coupling. The effect of ACh on IR was greatly potentiated by GLP-1 (10 nM). ACh caused a dose-dependent increase in [Ca2+]i at 0.1-1 microM or biphasic changes (an initial sharp increase in [Ca2+]i followed by a sustained phase of low [Ca2+]i) at 1-100 microM. The latter effects were observed in substrate-free medium or in the presence of 16.7 mM glucose. We conclude that SUR1 deletion depolarizes the beta-cells and markedly elevates basal [Ca2+]i. Elevated [Ca2+]i in turn sensitizes the beta-cells to the secretory effects of ACh and IBMX. Priming by the combination of high [Ca2+]i, ACh, and GLP-1 restores the defective glucose responsiveness, precluding the development of diabetes but not effectively enough to cause hyperinsulinemic hypoglycemia.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Acetilcolina/fisiologia , Glicemia/metabolismo , Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Cálcio/metabolismo , Glucagon/fisiologia , Peptídeo 1 Semelhante ao Glucagon , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Líquido Intracelular/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/deficiência , Fragmentos de Peptídeos/fisiologia , Inibidores de Fosfodiesterase/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização , Precursores de Proteínas/fisiologia , Receptores de Droga , Sistemas do Segundo Mensageiro/fisiologia , Transdução de Sinais/fisiologia , Receptores de Sulfonilureias
6.
Diabetes ; 52(2): 394-402, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12540613

RESUMO

In the present study, noninvasive (31)P and (23)Na(+)-nuclear magnetic resonance (NMR) technology and respirometry were used to compare the effect of high glucose (30 mmol/l) with the effect of the antidiabetic sulfonylurea (SU) compound glyburide (GLY) on energy metabolism, Na(+) flux, insulin, and cAMP release of continuously superfused beta-HC9 cells encapsulated in microscopic agarose beads. Both high glucose and GLY increased oxygen consumption in beta-HC9 cells (15-30%) with a maximal effect at 8 mmol/l for glucose and at 250 nmol/l for GLY. At the same time, insulin release from beta-cells increased by 15- and 25-fold with high glucose or GLY, respectively. The P-creatine (PCr) level was greatly increased and inorganic phosphate (P(i)) was decreased with 30 mmol/l glucose in contrast to the decreased level of PCr and increased P(i) with GLY. ATP levels remained unchanged during both interventions. Studies on isolated mitochondria of beta-HC9 cells showed that GLY added to mitochondria oxidizing glutamine or glutamate abolished the stimulation of respiration by ADP (state 3) meanwhile leaving state 3 respiration unchanged during oxidation of other substrates. Exposure of beta-HC9 cells to 5 mmol/l glucose decreased intracellular Na(+) levels monitored by (23)Na(+)-NMR spectroscopy and 30 mmol/l glucose resulted in a further decrease in cytosolic Na(+). In contrast, Na(+) increased when 1 micro mol/l GLY was added to the perfusate containing 5 mmol/l glucose. These data support the hypothesis that glucose activates the beta-cell through a "push mechanism" due to substrate pressure enhancing fuel flux, energy production, and extrusion of Na(+) from the cells in contrast to SU receptor (SUR)-1 inhibitors, which may modify intermediary and energy metabolism secondarily through a "pull mechanism" due to higher energy demand resulting from increased ion fluxes and the exocytotic work load.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Glucose/farmacologia , Glibureto/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Sódio/metabolismo , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Cinética , Espectroscopia de Ressonância Magnética/métodos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Perfusão/métodos , Fosfocreatina/metabolismo , Ratos , Ácido Succínico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...