Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Pediatr Diabetes ; 15(2): 127-34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23992543

RESUMO

CONTEXT: Glycemic control is limited by the barrier of hypoglycemia. Recurrent hypoglycemia impairs counterregulatory (CR) hormone responses to subsequent hypoglycemia. OBJECTIVE: To determine the glucagon and epinephrine responses to insulin-induced hypoglycemia in adolescents with recent-onset type 1 diabetes mellitus (T1DM). METHODS: We assessed the CR responses to hypoglycemia by performing a hyperinsulinemic (2.0 mU/kg/min), euglycemic (BG 90 mg/dL; 5.0 mmol/L)-hypoglycemic (BG 55 mg/dL; 3.0 mmol/L) clamp in 25 recent-onset (<1 yr duration) patients 9-18 yr old (mean ± SD: 13.4 ± 2.7) with T1DM and 16 non-diabetic controls 19-25 yr old (mean ± SD 23.3 ± 1.8). Twenty of the T1DM subjects were retested 1-yr (53 ± 3 wk) later. RESULTS: At the initial and 1-yr studies, peak glucagon (pGON) and incremental glucagon (ΔGON) during hypoglycemia were lower in the T1DM subjects [median pGON = 47 pg/mL (quartiles: 34, 72), ΔGON = 16 (4, 27) initially and pGON = 50 pg/mL (42, 70), ΔGON = 12 (9, 19) at 1-yr] than in controls [pGON = 93 pg/mL (60, 111); ΔGON = 38 pg/mL (19, 66), p = 0.01 and p = 0.004 for ΔGON at initial and 1-yr study, respectively]. In contrast, peak epinephrine (pEPI) and incremental epinephrine (ΔEPI) levels were similar in the T1DM (pEPI = 356 pg/mL (174, 797) and ΔEPI = 322 pg/mL (143, 781) initially and pEPI = 469 pg/mL (305, 595) and ΔEPI = 440 pg/mL (285, 574) at 1 yr) and in controls (pEPI = 383 pg/mL (329, 493) and ΔEPI = 336 pg/mL (298, 471) p = 0.97 and 0.21 for ΔEPI at initial and 1-yr study, respectively). CONCLUSIONS: Even within the first year of T1DM, glucagon responses to hypoglycemia are blunted but epinephrine responses are not, suggesting that the mechanisms involved in the loss of these hormonal responses, which are key components in pathophysiology of hypoglycemia-associated autonomic failure, are different.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Epinefrina/metabolismo , Glucagon/metabolismo , Hipoglicemia/metabolismo , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Adolescente , Adulto , Criança , Diabetes Mellitus Tipo 1/complicações , Progressão da Doença , Feminino , Técnica Clamp de Glucose , Humanos , Hipoglicemia/induzido quimicamente , Masculino , Adulto Jovem
3.
J Clin Invest ; 117(4): 868-70, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17404614

RESUMO

Hypoglycemia commonly causes brain fuel deprivation, resulting in functional brain failure, which can be corrected by raising plasma glucose concentrations. Rarely, profound hypoglycemia causes brain death that is not the result of fuel deprivation per se. In this issue of the JCI, Suh and colleagues use cell culture and in vivo rodent studies of glucose deprivation and marked hypoglycemia and provide evidence that hypoglycemic brain neuronal death is in fact increased by neuronal NADPH oxidase activation during glucose reperfusion (see the related article beginning on page 910). This finding suggests that, at least in the setting of profound hypoglycemia, therapeutic hyperglycemia should be avoided.


Assuntos
Morte Encefálica , Encefalopatias/etiologia , Transtornos Cognitivos/etiologia , Hipoglicemia/complicações , Glicemia/metabolismo , Humanos , Neurônios/patologia , Neurônios/fisiologia
4.
J Clin Invest ; 116(6): 1470-3, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16741570

RESUMO

A reduced sympathoadrenal response, induced by recent antecedent hypoglycemia, is the key feature of hypoglycemia-associated autonomic failure (HAAF) and, thus, the pathogenesis of iatrogenic hypoglycemia in diabetes. Understanding of the mechanism(s) of that reduced response awaits new insight into its basic molecular, cellular, organ, and whole-body physiology and pathophysiology in experimental models. In this issue of the JCI, McCrimmon and colleagues report that application of urocortin I (a corticotrophin-releasing factor receptor-2 agonist) to the ventromedial hypothalamus reduces the glucose counterregulatory response to hypoglycemia in rats (see the related article beginning on page 1723). Thus, hypothalamic urocortin I release during antecedent hypoglycemia is, among other possibilities, a potential mechanism of HAAF.


Assuntos
Glândulas Suprarrenais/fisiologia , Doenças do Sistema Nervoso Autônomo/fisiopatologia , Hormônio Liberador da Corticotropina/metabolismo , Diabetes Mellitus/fisiopatologia , Hipoglicemia/fisiopatologia , Animais , Doenças do Sistema Nervoso Autônomo/etiologia , Complicações do Diabetes/fisiopatologia , Glucagon/metabolismo , Humanos , Hipoglicemia/etiologia , Insulina/metabolismo , Ratos , Urocortinas , Núcleo Hipotalâmico Ventromedial/metabolismo
6.
Clin Sci (Lond) ; 114(9): 589-90, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18197838

RESUMO

Glucagon, in the setting of absolute or relative insulin deficiency, is thought to contribute to the pathogenesis of hyperglycaemia in diabetes, but much of the evidence is extrapolated from short-term studies to the long-term condition. In the present issue of Clinical Science, Li and co-workers report that infusion of glucagon raised fasting plasma glucose concentrations and impaired glucose tolerance over 4 weeks in mice, thus demonstrating a sustained glycaemic effect of hyperglucagonaemia. Nonetheless, compelling evidence that glucagon contributes to the pathogenesis of hyperglycaemia in diabetes awaits long-term selective reduction of glucagon secretion or action in humans.


Assuntos
Diabetes Mellitus/metabolismo , Glucagon/fisiologia , Hiperglicemia/metabolismo , Animais , Glicemia/metabolismo , Jejum , Humanos , Hipoglicemiantes/metabolismo , Hipoglicemiantes/uso terapêutico , Insulina/metabolismo
7.
Diabetes Care ; 40(12): 1641-1643, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29162584

RESUMO

The view that a hemoglobin A1c (A1C) level <7% (55 mmol/mol) is the accepted glycemic goal for most people with diabetes sometimes conflicts with the view that glycemic goals should be individualized and, thus, that somewhat higher A1C levels are appropriate for some, particularly many at risk for iatrogenic hypoglycemia because of treatment with insulin, a sulfonylurea, or a glinide. The relationship between A1C and chronic complications of diabetes is curvilinear, A1C is a relatively weak predictor of cardiovascular disease, and minor elevations of A1C above 7% have not been found to be associated with increased mortality. Iatrogenic hypoglycemia causes recurrent morbidity in diabetes and is sometimes fatal. In those at risk for hypoglycemia, a reasonable individualized glycemic goal is the lowest A1C that does not cause severe hypoglycemia and preserves awareness of hypoglycemia, preferably with little or no symptomatic or even asymptomatic hypoglycemia, at a given stage in the evolution of the individual's diabetes. A somewhat higher A1C level is appropriate in those who have previously experienced hypoglycemia or have potential high risk for hypoglycemia, have a long duration of diabetes, and have a short life expectancy, among other traits. Given the importance of severe hypoglycemia in selecting glycemic goals, it is proposed to expand the classification of severe hypoglycemia beyond a hypoglycemic event requiring assistance from another person to include a measured glucose concentration <50 mg/dL (2.8 mmol/L), a level associated with sudden death.


Assuntos
Glicemia/metabolismo , Doenças Cardiovasculares/epidemiologia , Diabetes Mellitus Tipo 2/epidemiologia , Hipoglicemia/sangue , Hipoglicemia/tratamento farmacológico , Hipoglicemia/epidemiologia , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/diagnóstico , Doença Crônica , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Gerenciamento Clínico , Seguimentos , Hemoglobinas Glicadas/metabolismo , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Morbidade , Compostos de Sulfonilureia/uso terapêutico
8.
Diabetes ; 66(12): 2964-2972, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28970283

RESUMO

Hypoglycemia is the most common complication of diabetes, causing morbidity and death. Recurrent hypoglycemia alters the cascade of physiological and behavioral responses that maintain euglycemia. The extent to which these responses are normally triggered by decreased whole-brain cerebral glucose metabolism (CMRglc) has not been resolved by previous studies. We measured plasma counterregulatory hormonal responses and whole-brain CMRglc (along with blood-to-brain glucose transport rates and brain glucose concentrations) with 1-[11C]-d-glucose positron emission tomography during hyperinsulinemic glucose clamps at nominal plasma glucose concentrations of 90, 75, 60, and 45 mg/dL (5.0, 4.2, 3.3, and 2.5 mmol/L) in 18 healthy young adults. Clear evidence of hypoglycemic physiological counterregulation was first demonstrated between 75 mg/dL (4.2 mmol/L) and 60 mg/dL (3.3 mmol/L) with increases in both plasma epinephrine (P = 0.01) and glucagon (P = 0.01). In contrast, there was no statistically significant change in CMRglc (P = 1.0) between 75 mg/dL (4.2 mmol/L) and 60 mg/dL (3.3 mmol/L), whereas CMRglc significantly decreased (P = 0.02) between 60 mg/dL (3.3 mmol/L) and 45 mg/dL (2.5 mmol/L). Therefore, the increased epinephrine and glucagon secretion with declining plasma glucose concentrations is not in response to a decrease in whole-brain CMRglc.


Assuntos
Encéfalo/metabolismo , Epinefrina/sangue , Glucagon/sangue , Glucose/metabolismo , Hipoglicemia/metabolismo , Adulto , Feminino , Humanos , Masculino , Tomografia por Emissão de Pósitrons
9.
J Clin Endocrinol Metab ; 91(6): 2087-92, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16492699

RESUMO

OBJECTIVE: We assessed four putative bedtime treatments in the prevention of nocturnal hypoglycemia in type 1 diabetes. RESEARCH DESIGN AND METHODS: Plasma glucose concentrations were measured every 15 min from 2200 h through 0700 h in 21 patients with type 1 diabetes (mean +/- sd HbA(1C) = 7.1 +/- 1.0%) on five occasions with, in random sequence, bedtime (2200 h) administration of 1) no treatment, 2) a snack, 3) the snack plus the alpha-glucosidase inhibitor acarbose, 4) an uncooked cornstarch bar, or 5) the beta(2)-adrenergic agonist terbutaline. RESULTS: In the absence of a bedtime treatment, 27% of the measured nocturnal plasma glucose concentrations were less than 70 mg/dl (3.9 mmol/liter) in 12 patients; 16, 6, and 1% were less than 60, less than 50, and less than 40 mg/dl (3.3, 2.8, and 2.2 mmol/liter), respectively. Neither the snack (without or with acarbose) nor cornstarch raised the mean nadir nocturnal glucose concentration or reduced the number of low glucose levels or the number of patients with low levels. Terbutaline raised the mean nadir nocturnal glucose concentration (mean +/- se, 127 +/- 11 vs. 75 +/- 9 mg/dl; P < 0.001), eliminated glucose levels less than 50 mg/dl (P = 0.038), reduced levels less than 60 mg/dl (P = 0.005) to one, and reduced levels less than 70 mg/dl (P = 0.001) to five (four at 2215 h, one at 2230 h). However, it also raised glucose levels the following morning. CONCLUSIONS: Nocturnal hypoglycemia is common in aggressively treated type 1 diabetes. Bedtime administration of a conventional snack or of uncooked cornstarch does not prevent it. That of terbutaline prevents nocturnal hypoglycemia but causes hyperglycemia the following morning. The efficacy of a lower dose of terbutaline remains to be determined.


Assuntos
Glicemia/análise , Ritmo Circadiano , Diabetes Mellitus Tipo 1/sangue , Hipoglicemia/prevenção & controle , Acarbose/uso terapêutico , Adulto , Pressão Sanguínea , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Frequência Cardíaca , Humanos , Masculino , Terbutalina/uso terapêutico
10.
Diabetes ; 54(12): 3592-601, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16306382

RESUMO

Iatrogenic hypoglycemia is a problem for people with diabetes. It causes recurrent morbidity, and sometimes death, as well as a vicious cycle of recurrent hypoglycemia, precluding maintenance of euglycemia over a lifetime of diabetes. Improved therapeutic approaches that will minimize both hypo- and hyperglycemia will be based on insight into the pathophysiology of glucoregulation, specifically glucose counterregulation, in insulin-deficient (type 1 and advanced type 2) diabetes. In such patients, hypoglycemia is the result of the interplay of relative or absolute therapeutic insulin excess and compromised physiological (the syndrome of defective glucose counterregulation) and behavioral (the syndrome of hypoglycemia unawareness) defenses against falling plasma glucose concentrations. The concept of hypoglycemia-associated autonomic failure (HAAF) in diabetes posits that recent antecedent iatrogenic hypoglycemia causes both defective glucose counterregulation (by reducing epinephrine responses to a given level of subsequent hypoglycemia in the setting of absent decrements in insulin and absent increments in glucagon) and hypoglycemia unawareness (by reducing sympathoadrenal and the resulting neurogenic symptom responses to a given level of subsequent hypoglycemia) and thus a vicious cycle of recurrent hypoglycemia. The clinical impact of HAAF is well established in type 1 diabetes; it also affects those with advanced type 2 diabetes. It is now known to be largely reversible, by as little as 2-3 weeks of scrupulous avoidance of hypoglycemia, in most affected patients. However, the mechanisms of HAAF and its component syndromes are largely unknown. Loss of the glucagon secretory response, a key feature of defective glucose counterregulation, is plausibly explained by insulin deficiency, specifically loss of the decrement in intraislet insulin that normally signals glucagon secretion as glucose levels fall. Reduced neurogenic symptoms, a key feature of hypoglycemia unawareness, are largely the result of reduced sympathetic neural responses to falling glucose levels. The mechanism by which hypoglycemia shifts the glycemic thresholds for sympathoadrenal activation to lower plasma glucose concentrations, the key feature of both components of HAAF, is not known. It does not appear to be the result of the release of a systemic mediator (e.g., cortisol, epinephrine) during antecedent hypoglycemia or of increased blood-to-brain glucose transport (although increased transport of alternative fuels is conceivable). It is likely the result of alterations of brain metabolism. Although there is an array of clues, the specific alteration remains to be identified. While the research focus has been largely on the hypothalamus, hypoglycemia is now known to activate widespread brain regions, including the medial prefrontal cortex. The possibility that HAAF could be the result of posthypoglycemic brain glycogen supercompensation has also been raised. Finally, there appear to be diverse causes of HAAF. In addition to recent antecedent hypoglycemia, these include exercise- and sleep-related HAAF. Clearly, a unifying mechanism of HAAF would need to incorporate these causes as well. Pending the prevention and cure of diabetes, critical fundamental, translational, and outcomes research is needed if we are to eliminate hypoglycemia from the lives of people affected by diabetes.


Assuntos
Doenças do Sistema Nervoso Autônomo/fisiopatologia , Diabetes Mellitus/fisiopatologia , Hipoglicemia/complicações , Diabetes Mellitus/sangue , Humanos , Síndrome
11.
Diabetes ; 54(3): 757-64, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15734853

RESUMO

The intraislet insulin hypothesis for the signaling of the glucagon secretory response to hypoglycemia states that a decrease in arterial glucose --> a decrease in beta-cell insulin secretion --> a decrease in tonic alpha-cell inhibition by insulin --> an increase in alpha-cell glucagon secretion. To test this hypothesis in humans, a hyperinsulinemic- euglycemic ( approximately 5.0 mmol/l [90 mg/dl] x 2 h) and then a hypoglycemic ( approximately 3.0 mmol/l [55 mg/dl] x 2 h) clamp was performed in 14 healthy young adults on two occasions, once with oral administration of the ATP-sensitive potassium channel agonist diazoxide to selectively suppress baseline insulin secretion and once with the administration of a placebo. The decrement in plasma C-peptide during the induction of hypoglycemia was reduced by approximately 50% in the diazoxide clamps (from 0.3 +/- 0.0 to 0.1 +/- 0.0 nmol/l [0.8 +/- 0.1 to 0.4 +/- 0.1 ng/ml]) compared with the placebo clamps (from 0.4 +/- 0.0 to 0.1 +/- 0.0 nmol/l [1.2 +/- 0.1 to 0.4 +/- 0.1 ng/ml]) (P = 0.0015). This reduction of the decrement in intraislet insulin during induction of hypoglycemia caused an approximately 50% reduction (P = 0.0010) of the increase in plasma glucagon in the diazoxide clamps (from 29 +/- 3 to 35 +/- 2 pmol/l [102 +/- 9 to 123 +/- 8 pg/ml]) compared with the placebo clamps (from 28 +/- 2 to 43 +/- 5 pmol/l [98 +/- 7 to 151 +/- 16 pg/ml]). Baseline glucagon levels, the glucagon response to intravenous arginine, and the autonomic (adrenomedullary, sympathetic neural, and parasympathetic neural) responses to hypoglycemia were not altered by diazoxide. These data indicate that a decrease in intraislet insulin is a signal for the glucagon secretory response to hypoglycemia in healthy humans. The absence of that signal plausibly explains the loss of the glucagon response to falling plasma glucose concentrations, a key feature of the pathogenesis of iatrogenic hypoglycemia, in insulin-deficient (type 1 and advanced type 2) diabetes.


Assuntos
Glucagon/fisiologia , Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Adulto , Diazóxido/farmacologia , Feminino , Glucagon/metabolismo , Hormônio do Crescimento/sangue , Humanos , Hidrocortisona/sangue , Hipoglicemia/sangue , Hipoglicemia/fisiopatologia , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Transdução de Sinais
12.
Prog Brain Res ; 153: 361-5, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16876586

RESUMO

Iatrogenic hypoglycemia, the limiting factor in the glycemic management of diabetes, causes recurrent morbidity (and sometimes death), precludes maintenance of euglycemia over a lifetime of diabetes and causes a vicious cycle of recurrent hypoglycemia. In insulin deficient - T1DM and advanced T2DM - diabetes hypoglycemia is the result of the interplay of therapeutic insulin excess and compromised physiological (defective glucose counterregulation) and behavioral (hypoglycemia unawareness) defenses against falling plasma glucose concentrations. The concept of hypoglycemia-associated autonomic failure (HAAF) in diabetes posits that recent antecedent hypoglycemia causes both defective glucose counterregulation (by reducing epinephrine responses in the setting of absent insulin and glucagon responses) and hypoglycemia unawareness (by reducing sympathoadrenal and the resulting neurogenic symptom responses) and thus a vicious cycle of recurrent hypoglycemia. The clinical impact of HAAF-including its reversal by avoidance of hypoglycemia-is well established, but its mechanisms are largely unknown. Loss of the glucagon response, a key feature of defective glucose counterregulation, is plausibly attributed to insulin deficiency, specifically loss of the decrement in intraislet insulin that normally signals glucagon secretion as glucose levels fall. Reduced neurogenic symptoms, a key feature of hypoglycemia unawareness, are largely the result of reduced sympathetic neural responses to falling glucose levels. The mechanism(s) by which hypoglycemia shifts the glycemic thresholds for sympathoadrenal activation to lower plasma glucose concentrations, the key feature of both components of HAAF, is not known. It does not appear to be the result of the release of a systemic mediator such as cortisol or epinephrine during antecedent hypoglycemia or of increased blood-to-brain glucose transport. It is likely the result of an as yet to be identified alteration of brain metabolism. While the research focus has been largely on the hypothalamus, hypoglycemia is known to activate widespread brain regions including the medial prefrontal cortex. The possibility of post-hypoglycemic brain glycogen supercompensation has also been raised. Finally, a unifying mechanism of HAAF would need to incorporate the effects of sleep and antecedent exercise which produce a phenomenon similar to hypoglycemia induced HAAF.


Assuntos
Ritmo Circadiano/fisiologia , Diabetes Mellitus/fisiopatologia , Hipoglicemia/fisiopatologia , Animais , Doenças do Sistema Nervoso Autônomo/etiologia , Humanos , Hipoglicemia/etiologia
13.
Metabolism ; 55(1): 78-83, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16324923

RESUMO

Severe hypoglycemia, the most serious side effect of sulfonylurea therapy, has been reported to occur more frequently with glyburide than glimepiride. The present studies were undertaken to test the hypothesis that a differential effect on glucagon secretion may be involved. We performed hyperinsulinemic hypoglycemic (approximately 2.5 mmol/L) clamps in 16 healthy volunteers who received in randomized order placebo, glyburide (10 mg), and glimepiride (4 mg) just before beginning the insulin infusion and measured plasma glucagon, insulin, C-peptide, glucagon, epinephrine, cortisol, and growth hormone levels during the clamp and during a 3-hour recovery period after discontinuation of the insulin infusion. Neither sulfonylurea altered glucagon responses or those of other counterregulatory hormones (except cortisol) during the clamp. However, glyburide delayed plasma glucose recovery from hypoglycemia (plasma glucose at end of recovery period: control, 4.9 +/- 0.2 mmol/L; glyburide, 3.7 +/- 0.2 mmol/L; P = .0001; glimepiride, 4.5 +/- 0.2 mmol/L; P = .08). Despite lower plasma glucose levels, glyburide stimulated insulin secretion during this period (0.89 +/- 0.13 vs 1.47 +/- 0.15 pmol x kg(-1) x min(-1), control vs glyburide; P = .001), whereas glimepiride did not (P = .08). Short-term administration of glyburide or glimepiride did not alter glucagon responses during hypoglycemia. In contrast, during recovery from hypoglycemia, glyburide but not glimepiride inappropriately stimulates insulin secretion at low plasma glucose levels. This differential effect on insulin secretion may be an important factor in explaining why glyburide causes severe hypoglycemia more frequently than glimepiride.


Assuntos
Glucose/metabolismo , Glibureto/farmacologia , Hipoglicemia/tratamento farmacológico , Hipoglicemiantes/farmacologia , Compostos de Sulfonilureia/farmacologia , Adulto , Glicemia/metabolismo , Peptídeo C/sangue , Epinefrina/sangue , Feminino , Hormônios/sangue , Hormônio do Crescimento Humano/sangue , Humanos , Hidrocortisona/sangue , Hipoglicemia/sangue , Insulina/sangue , Cinética , Masculino
14.
Metabolism ; 55(5): 593-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16631434

RESUMO

In type 2 diabetes mellitus (T2DM), little is known about hormonal responses to hypoglycemia. In particular, beta-cell responses to hypoglycemia have not been carefully investigated and potentially because of confounding factors or insufficient power, conflicting data have been obtained regarding growth hormone responses. We therefore compared hormonal responses including rates of insulin secretion during a 2-hour hyperinsulinemic hypoglycemic clamp in a relatively large number of nondiabetic (n=21) and moderately insulin-deficient subjects with T2DM (homeostasis model assessment of beta-cell function [HOMA-%B], 751+/-160 vs 1144+/-83 [pmol/L]/[mmol/L], P<.04) (n=14) matched for age, sex, and body mass index. Subjects with T2DM were excluded for antecedent hypoglycemia, and baseline glycemia was controlled by a variable infusion of insulin overnight. Although both groups of subjects had indistinguishable plasma glucose levels at baseline and virtually identical levels of plasma insulin and glucose throughout the hypoglycemic clamp, insulin secretion decreased more slowly in the subjects with T2DM. The time required for insulin secretion to decline to half its baseline level was markedly increased (38.9+/-4.9 vs 22.3+/-1.3 minutes [SD], P<.01), and insulin secretion decreased to a lesser extent (-0.79+/-0.17 vs -1.51+/-0.09 [pmol/L]/kg per minute, P<.002). Moreover, responses of glucagon (28.3+/-7.3 vs 52.8+/-7.0 ng/L, P<.05) and growth hormone (2.9+/-0.8 vs 6.3+/-0.9 ng/mL, P<.04) were reduced in the subjects with T2DM, whereas responses of epinephrine, norepinephrine, and cortisol were similar to those in nondiabetic subjects (all P>0.6). We conclude that multiple defects exist in hormonal responses to hypoglycemia in T2DM with moderate beta-cell failure. These include delayed and reduced decreases in insulin secretion, and impaired increases of plasma glucagon and growth hormone.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Hormônio do Crescimento Humano/metabolismo , Hipoglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Adulto , Proteína C-Reativa/metabolismo , Diabetes Mellitus Tipo 2/sangue , Epinefrina/sangue , Epinefrina/metabolismo , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Feminino , Glucagon/sangue , Glucagon/metabolismo , Hormônio do Crescimento Humano/sangue , Humanos , Hidrocortisona/sangue , Hidrocortisona/metabolismo , Insulina/sangue , Secreção de Insulina , Masculino , Norepinefrina/sangue , Norepinefrina/metabolismo , Análise de Regressão
15.
Diabetes Care ; 28(5): 1124-31, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15855577

RESUMO

OBJECTIVE: Animal and in vitro studies indicate that a decrease in beta-cell insulin secretion, and thus a decrease in tonic alpha-cell inhibition by intraislet insulin, may be an important factor for the increase in glucagon secretion during hypoglycemia. However, in humans this role of decreased intraislet insulin is still unclear. RESEARCH DESIGN AND METHODS: We studied glucagon responses to hypoglycemia in 14 nondiabetic subjects on two separate occasions. On both occasions, insulin was infused from 0 to 120 min to induce hypoglycemia. On one occasion, somatostatin was infused from -60 to 60 min to suppress insulin secretion, so that the decrement in intraislet insulin during the final 60 min of hypoglycemia would be reduced. On the other occasion, subjects received an infusion of normal saline instead of the somatostatin. RESULTS: During the 2nd h of the insulin infusion, when somatostatin or saline was no longer being infused, plasma glucose ( approximately 2.6 mmol/l) and insulin levels ( approximately 570 pmol/l) were comparable in both sets of experiments (both P > 0.4). In the saline experiments, insulin secretion remained unchanged from baseline (-90 to -60 min) before insulin infusion and decreased from 1.20 +/- 0.12 to 0.16 +/- 0.04 pmol . kg(-1) . min(-1) during insulin infusion (P < 0.001). However, in the somatostatin experiments, insulin secretion decreased from 1.18 +/- 0.12 pmol . kg(-1) . min(-1) at baseline to 0.25 +/- 0.09 pmol . kg(-1) . min(-1) before insulin infusion so that it did not decrease further during insulin infusion (-0.12 +/- 0.10 pmol . kg(-1) . min(-1), P = 0.26) indicating the complete lack of a decrement in intraislet insulin during hypoglycemia. This was associated with approximately 30% lower plasma glucagon concentrations (109 +/- 7 vs. 136 +/- 9 pg/ml, P < 0.006) and increments in plasma glucagon above baseline (41 +/- 8 vs. 67 +/- 11 pg/ml, P < 0.008) during the last 15 min of the hypoglycemic clamp. In contrast, increases in plasma growth hormone were approximately 70% greater during hypoglycemia after somatostatin infusion (P < 0.007), suggesting that to some extent the increases in plasma glucagon might have reflected a rebound in glucagon secretion. CONCLUSIONS: These results provide direct support for the intraislet insulin hypothesis in humans. However, the exact extent to which a decrement in intraislet insulin accounts for the glucagon responses to hypoglycemia remains to be established.


Assuntos
Glucagon/sangue , Hipoglicemia/metabolismo , Hipoglicemiantes/sangue , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Adulto , Glicemia , Peptídeo C/sangue , Epinefrina/sangue , Feminino , Glucagon/metabolismo , Hormônio do Crescimento Humano/sangue , Humanos , Hidrocortisona/sangue , Hipoglicemia/induzido quimicamente , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Insulina/metabolismo , Secreção de Insulina , Masculino , Somatostatina/administração & dosagem
16.
Diabetes Care ; 28(11): 2691-6, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16249541

RESUMO

OBJECTIVE: In advanced beta-cell failure, counterregulatory glucagon responses may be impaired due to a reduced decrement in insulin secretion during the development of hypoglycemia. The present studies were therefore undertaken to test the hypothesis that these may be improved by increasing this decrement in insulin secretion. RESEARCH DESIGN AND METHODS: Twelve subjects with type 2 diabetes who have been insulin requiring were studied as a model of advanced beta-cell failure. Glucagon responses were examined during a 90-min hypoglycemic clamp (approximately 2.8 mmol/l) on two separate occasions. On one occasion, tolbutamide was infused for 2 h before the clamp so that the decrement in insulin secretion during the induction of hypoglycemia would be increased. On the other occasion, normal saline was infused as a control. RESULTS: Before the hypoglycemic clamp, infusion of tolbutamide increased insulin secretion approximately 1.9-fold (P < 0.001). However, during hypoglycemia, insulin secretion decreased to similar rates on both occasions (P = 0.31) so that its decrement was approximately twofold greater following the tolbutamide infusion (1.63 +/- 0.20 vs. 0.81 +/- 0.17 pmol x kg(-1) x min(-1), P < 0.001). This was associated with more than twofold-greater glucagon responses (42 +/- 11 vs. 19 +/- 8 ng/l, P < 0.002) during the hypoglycemic clamp but unaltered glucagon responses to intravenous arginine immediately thereafter (449 +/- 50 vs. 453 +/- 50 ng/l, P = 0.78). CONCLUSIONS: Increasing the decrement in insulin secretion during the development of hypoglycemia improves counterregulatory glucagon responses in advanced beta-cell failure. These findings further support the concept that the impaired counterregulatory glucagon responses in advanced beta-cell failure may at least partially be due to a reduced decrement in insulin secretion.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Glucagon/metabolismo , Hipoglicemia/metabolismo , Hipoglicemiantes/sangue , Insulina/sangue , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Glucagon/sangue , Técnica Clamp de Glucose , Humanos , Hipoglicemia/induzido quimicamente , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/metabolismo , Insulina/administração & dosagem , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Tolbutamida/uso terapêutico
17.
Diabetes ; 52(5): 1195-203, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12716752

RESUMO

Given that iatrogenic hypoglycemia often occurs during the night in people with type 1 diabetes, we tested the hypothesis that physiological, and the resulting behavioral, defenses against developing hypoglycemia-already compromised by absent glucagon and attenuated epinephrine and neurogenic symptom responses-are further compromised during sleep in type 1 diabetes. To do so, we studied eight adult patients with uncomplicated type 1 diabetes and eight matched nondiabetic control subjects with hyperinsulinemic stepped hypoglycemic clamps (glucose steps of approximately 85, 75, 65, 55, and 45 mg/dl) in the morning (0730-1230) while awake and at night (2100-0200) while awake throughout and while asleep from 0000 to 0200 in random sequence. Plasma epinephrine (P = 0.0010), perhaps norepinephrine (P = 0.0838), and pancreatic polypeptide (P = 0.0034) responses to hypoglycemia were reduced during sleep in diabetic subjects (the final awake versus asleep values were 240 +/- 86 and 85 +/- 47, 205 +/- 24 and 148 +/- 17, and 197 +/- 45 and 118 +/- 31 pg/ml, respectively), but not in the control subjects. The diabetic subjects exhibited markedly reduced awakening from sleep during hypoglycemia. Sleep efficiency (percent time asleep) was 77 +/- 18% in the diabetic subjects, but only 26 +/- 8% (P = 0.0109) in the control subjects late in the 45-mg/dl hypoglycemic steps. We conclude that autonomic responses to hypoglycemia are reduced during sleep in type 1 diabetes, and that, probably because of their reduced sympathoadrenal responses, patients with type 1 diabetes are substantially less likely to be awakened by hypoglycemia. Thus both physiological and behavioral defenses are further compromised during sleep. This sleep-related hypoglycemia-associated autonomic failure, in the context of imperfect insulin replacement, likely explains the high frequency of nocturnal hypoglycemia in type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Neuropatias Diabéticas/fisiopatologia , Hipoglicemia/fisiopatologia , Fases do Sono/fisiologia , Sono/fisiologia , Vigília/fisiologia , Glicemia/metabolismo , Peptídeo C/sangue , Ritmo Circadiano , Diabetes Mellitus Tipo 1/sangue , Neuropatias Diabéticas/sangue , Humanos , Hipoglicemia/etiologia , Insulina/sangue , Valores de Referência , Sono REM/fisiologia
18.
Diabetes ; 52(8): 2083-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12882926

RESUMO

The concept of hypoglycemia-associated autonomic failure (HAAF) in diabetes posits that recent antecedent iatrogenic hypoglycemia causes both defective glucose counterregulation (by reducing the epinephrine response in the setting of an absent glucagon response) and hypoglycemia unawareness (by reducing the autonomic-sympathetic neural and adrenomedullary response and the resulting neurogenic [autonomic] symptom responses) and thus causes a vicious cycle of recurrent hypoglycemia. To assess the suggestion that it is the cortisol response to antecedent hypoglycemia that mediates HAAF, we tested the hypothesis that plasma cortisol elevations during euglycemia that are comparable to those that occur during hypoglycemia reduce sympathoadrenal and neurogenic symptom responses to subsequent hypoglycemia. To do this, 12 healthy subjects were studied with hyperinsulinemic-stepped hypoglycemic clamps the day after saline or cortisol (1.3 +/- 0.2 micro g. kg(-1) x min(-1)) infusions from 0930 to 1200 and from 1330 to 1600. Compared with saline, antecedent cortisol elevations did not reduce the sympathoadrenal (e.g., final plasma epinephrine levels of 674 +/- 84 vs. 606 +/- 80 pg/ml and final plasma norepinephrine levels of 332 +/- 26 vs. 304 +/- 26 pg/ml) or neurogenic symptom (e.g., final scores of 9.3 +/- 1.1 vs. 13.2 +/- 1.3) responses to subsequent hypoglycemia. Thus, these data do not support the suggestion that cortisol mediates HAAF.


Assuntos
Anti-Inflamatórios/sangue , Doenças do Sistema Nervoso Autônomo/sangue , Doenças do Sistema Nervoso Autônomo/etiologia , Hidrocortisona/sangue , Hipoglicemia/sangue , Adulto , Anti-Inflamatórios/administração & dosagem , Peptídeo C/sangue , Neuropatias Diabéticas/sangue , Neuropatias Diabéticas/etiologia , Epinefrina/sangue , Feminino , Técnica Clamp de Glucose , Humanos , Hidrocortisona/administração & dosagem , Hiperinsulinismo/sangue , Masculino , Norepinefrina/sangue , Cloreto de Sódio/administração & dosagem
19.
Diabetes ; 51(4): 958-65, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11916913

RESUMO

Because absence of the glucagon response to falling plasma glucose concentrations plays a key role in the pathogenesis of iatrogenic hypoglycemia in patients with insulin-deficient diabetes and the mechanism of this defect is unknown, and given evidence in experimental animals that a decrease in intraislet insulin is a signal to increased glucagon secretion, we examined the role of endogenous insulin in the physiological glucagon response to hypoglycemia. We tested the hypothesis that intraislet hyperinsulinemia prevents the glucagon response to hypoglycemia despite an intact autonomic-adrenomedullary, sympathetic neural, and parasympathetic neural-response and a low alpha-cell glucose concentration. Twelve healthy young adults were studied on three separate occasions. Insulin was infused in hourly steps in relatively low doses (1.5, 3.0, 4.5, and 6.0 pmol.kg(-1).min(-1)) from 60 through 300 min on all three occasions. Plasma glucose levels were clamped at euglycemia ( approximately 5.0 mmol/l, approximately 90 mg/dl) on one occasion and at hourly steps of approximately 4.7, 4.2, 3.6, and 3.0 mmol/l ( approximately 85, 75, 65, and 55 mg/dl) from 60 through 300 min on the other two occasions. On one of the latter occasions, the beta-cell secretagogue tolbutamide was infused in a dose of 1.0 g/h from 60 through 300 min. Hypoglycemia with tolbutamide infusion, compared with similar hypoglycemia alone, was associated with higher (P < 0.0001) C-peptide levels (final values of 1.0 +/- 0.2 vs. 0.1 +/- 0.0 nmol/l), higher (P < 0.0001) rates of insulin secretion (final values of 198 +/- 60 vs. 15 +/- 4 pmol/min), and higher (P < 0.0001) insulin levels (final values of 325 +/- 30 vs. 245 +/- 20 pmol/l) as expected. The glucagon response to hypoglycemia was prevented during tolbutamide infusion (P < 0.0001). Glucagon levels were 17 +/- 1 pmol/l at baseline on both occasions, 14 +/- 1 vs. 15 +/- 1 pmol/l, respectively, during the initial hyperinsulinemic euglycemia, and 15 +/- 1 vs. 22 +/- 2 pmol/l, respectively, during hypoglycemia with and without tolbutamide infusion. Autonomic-adrenomedullary (plasma epinephrine), sympathetic neural (plasma norepinephrine), and parasympathetic neural (plasma pancreatic polypeptide)-responses to hypoglycemia were not reduced during tolbutamide infusion. We conclude that intraislet hyperinsulinemia prevents the glucagon response to hypoglycemia despite an intact autonomic response and a low alpha-cell glucose concentration.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Glucagon/metabolismo , Hiperinsulinismo/fisiopatologia , Insulina/metabolismo , Insulina/farmacologia , Ilhotas Pancreáticas/metabolismo , Adulto , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Epinefrina/sangue , Feminino , Glucagon/antagonistas & inibidores , Glucagon/sangue , Técnica Clamp de Glucose , Hormônio do Crescimento Humano/sangue , Humanos , Hidrocortisona/sangue , Infusões Intravenosas , Insulina/administração & dosagem , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Cinética , Masculino , Norepinefrina/sangue , Polipeptídeo Pancreático/sangue , Fatores de Tempo
20.
Diabetes ; 51(3): 724-33, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11872673

RESUMO

We tested the hypotheses that the glucagon response to hypoglycemia is reduced in patients who are approaching the insulin-deficient end of the spectrum of type 2 diabetes and that recent antecedent hypoglycemia shifts the glycemic thresholds for autonomic (including adrenomedullary epinephrine) and symptomatic responses to hypoglycemia to lower plasma glucose concentrations in type 2 diabetes. Hyperinsulinemic stepped hypoglycemic clamps (85, 75, 65, 55, and 45 mg/dl steps) were performed on two consecutive days, with an additional 2 h of hypoglycemia (50 mg/dl) in the afternoon of the first day, in 13 patients with type 2 diabetes---7 treated with oral hypoglycemic agents (OHA R(X); mean [+/- SD] HbA(1c) 8.6 +/- 1.1%) and 6 requiring therapy with insulin for an average of 5 years and with reduced C-peptide levels (insulin R(X), HbA(1c) 7.5 +/- 0.7%)---and 15 nondiabetic control subjects. The glucagon response to hypoglycemia was virtually absent (P = 0.0252) in the insulin-deficient type 2 diabetic patients (insulin R(X) mean [+/- SE] final values of 52 plus minus 16 vs. 93 plus minus 15 pg/ml in control subjects and 98 +/- 16 pg/ml in type 2 diabetic patients, OHA R(X) on day 1). Glucagon (P = 0.0015), epinephrine (P = 0.0002), and norepinephrine (P = 0.0138) responses and neurogenic (P = 0.0149) and neuroglycopenic (P = 0.0015) symptom responses to hypoglycemia were reduced on day 2 after hypoglycemia on day 1 in type 2 diabetic patients; these responses were not eliminated, but their glycemic thresholds were shifted to lower plasma glucose concentrations. In addition, the glycemic thresholds for these responses were at higher-than-normal plasma glucose concentrations (P = 0.0082, 0.0028, 0.0023, and 0.0182, respectively) at baseline (day 1) in OHA R(X) type 2 diabetic patients, with relatively poorly controlled diabetes. Because the glucagon response to falling plasma glucose levels is virtually absent and the glycemic thresholds for autonomic and symptomatic responses to hypoglycemia are shifted to lower glucose concentrations by recent antecedent hypoglycemia, patients with advanced type 2 diabetes, like those with type 1 diabetes, are at risk for hypoglycemia-associated autonomic failure and the resultant vicious cycle of recurrent iatrogenic hypoglycemia.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Hipoglicemia/fisiopatologia , Adulto , Glicemia/análise , Peptídeo C/sangue , Diabetes Mellitus Tipo 2/sangue , Epinefrina/sangue , Feminino , Glucagon/sangue , Técnica Clamp de Glucose , Hemoglobinas Glicadas/análise , Humanos , Insulina/sangue , Cinética , Masculino , Pessoa de Meia-Idade , Norepinefrina/sangue
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa