Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Diabetologia ; 63(10): 2086-2094, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32894319

RESUMEN

Increasing evidence suggests that, although pancreatic islets can function autonomously to detect and respond to changes in the circulating glucose level, the brain cooperates with the islet to maintain glycaemic control. Here, we review the role of the central and autonomic nervous systems in the control of the endocrine pancreas, including mechanisms whereby the brain senses circulating blood glucose levels. We also examine whether dysfunction in these systems might contribute to complications of type 1 diabetes and the pathogenesis of type 2 diabetes. Graphical abstract.


Asunto(s)
Sistema Nervioso Autónomo/metabolismo , Glucemia/metabolismo , Sistema Nervioso Central/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/inervación , Animales , Sistema Nervioso Autónomo/fisiopatología , Sistema Nervioso Central/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Células Receptoras Sensoriales
2.
Am J Physiol Endocrinol Metab ; 319(6): E1074-E1083, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33044845

RESUMEN

This study aimed to investigate the contributions of two factors potentially impairing glucagon response to insulin-induced hypoglycemia (IIH) in insulin-deficient diabetes: 1) loss of paracrine disinhibition by intra-islet insulin and 2) defects in the activation of the autonomic inputs to the islet. Plasma glucagon responses during hyperinsulinemic-hypoglycemic clamps ([Formula: see text]40 mg/dL) were assessed in dogs with spontaneous diabetes (n = 13) and in healthy nondiabetic dogs (n = 6). Plasma C-peptide responses to intravenous glucagon were measured to assess endogenous insulin secretion. Plasma pancreatic polypeptide, epinephrine, and norepinephrine were measured as indices of parasympathetic and sympathoadrenal autonomic responses to IIH. In 8 of the 13 diabetic dogs, glucagon did not increase during IIH (diabetic nonresponder [DMN]; ∆ = -6 ± 12 pg/mL). In five other diabetic dogs (diabetic responder [DMR]), glucagon responses (∆ = +26 ± 12) were within the range of nondiabetic control dogs (∆ = +27 ± 16 pg/mL). C-peptide responses to intravenous glucagon were absent in diabetic dogs. Activation of all three autonomic responses were impaired in DMN dogs but remained intact in DMR dogs. Each of the three autonomic responses to IIH was positively correlated with glucagon responses across the three groups. The study conclusions are as follows: 1) Impairment of glucagon responses in DMN dogs is not due to generalized impairment of α-cell function. 2) Loss of tonic inhibition of glucagon secretion by insulin is not sufficient to produce loss of the glucagon response; impairment of autonomic activation is also required. 3) In dogs with major ß-cell function loss, activation of the autonomic inputs is sufficient to mediate an intact glucagon response to IIH.NEW & NOTEWORTHY In dogs with naturally occurring, insulin-dependent (C-peptide negative) diabetes mellitus, impairment of glucagon responses is not due to generalized impairment of α-cell function. Loss of tonic inhibition of glucagon secretion by insulin is not sufficient, by itself, to produce loss of the glucagon response. Rather, impaired activation of the parasympathetic and sympathoadrenal autonomic inputs to the pancreas is also required. Activation of the autonomic inputs to the pancreas is sufficient to mediate an intact glucagon response to insulin-induced hypoglycemia in dogs with naturally occurring diabetes mellitus. These results have important implications that include leading to a greater understanding and insight into the pathophysiology, prevention, and treatment of hypoglycemia during insulin treatment of diabetes in companion dogs and in human patients.


Asunto(s)
Sistema Nervioso Autónomo/efectos de los fármacos , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/veterinaria , Enfermedades de los Perros/metabolismo , Glucagón/farmacología , Hipoglucemia/inducido químicamente , Hipoglucemia/metabolismo , Hipoglucemiantes , Insulina , Animales , Glucemia/metabolismo , Péptido C/metabolismo , Perros , Epinefrina/sangre , Células Secretoras de Glucagón/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Células Secretoras de Insulina/efectos de los fármacos , Norepinefrina/sangre , Polipéptido Pancreático/metabolismo
3.
Diabetologia ; 59(10): 2058-67, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27342407

RESUMEN

This review outlines the current state of knowledge regarding a unique neural defect of the pancreatic islet in autoimmune diabetes, one that we have termed early sympathetic islet neuropathy (eSIN). We begin with the findings that a majority of islet sympathetic nerves are lost near the onset of type 1, but not type 2, diabetes and that this nerve loss is restricted to the islet. We discuss later work demonstrating that while the loss of islet sympathetic nerves and the loss of islet beta cells in type 1 diabetes both require infiltration of the islet by lymphocytes, their respective mechanisms of tissue destruction differ. Uniquely, eSIN requires the activation of a specific neurotrophin receptor and we propose two possible pathways for activation of this receptor during the immune attack on the islet. We also outline what is known about the functional consequences of eSIN, focusing on impairment of sympathetically mediated glucagon secretion and its application to the clinical problem of insulin-induced hypoglycaemia. Finally, we offer our view on the important remaining questions regarding this unique neural defect.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/patología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología , Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/inmunología , Neuropatías Diabéticas/inmunología , Glucagón/metabolismo , Humanos , Factores de Crecimiento Nervioso/metabolismo , Sistema Nervioso Simpático/inmunología
4.
Am J Physiol Endocrinol Metab ; 309(3): E246-55, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26037249

RESUMEN

Short-term hyperglycemia suppresses superior cervical ganglia neurotransmission. If this ganglionic dysfunction also occurs in the islet sympathetic pathway, sympathetically mediated glucagon responses could be impaired. Our objectives were 1) to test for a suppressive effect of 7 days of streptozotocin (STZ) diabetes on celiac ganglia (CG) activation and on neurotransmitter and glucagon responses to preganglionic nerve stimulation, 2) to isolate the defect in the islet sympathetic pathway to the CG itself, and 3) to test for a protective effect of the WLD(S) mutation. We injected saline or nicotine in nondiabetic and STZ-diabetic rats and measured fos mRNA levels in whole CG. We electrically stimulated the preganglionic or postganglionic nerve trunk of the CG in nondiabetic and STZ-diabetic rats and measured portal venous norepinephrine and glucagon responses. We repeated the nicotine and preganglionic nerve stimulation studies in nondiabetic and STZ-diabetic WLD(S) rats. In STZ-diabetic rats, the CG fos response to nicotine was suppressed, and the norepinephrine and glucagon responses to preganglionic nerve stimulation were impaired. In contrast, the norepinephrine and glucagon responses to postganglionic nerve stimulation were normal. The CG fos response to nicotine, and the norepinephrine and glucagon responses to preganglionic nerve stimulation, were normal in STZ-diabetic WLD(S) rats. In conclusion, short-term hyperglycemia's suppressive effect on nicotinic acetylcholine receptors of the CG impairs sympathetically mediated glucagon responses. WLD(S) rats are protected from this dysfunction. The implication is that this CG dysfunction may contribute to the impaired glucagon response to insulin-induced hypoglycemia seen early in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/fisiopatología , Regulación hacia Abajo , Ganglios Simpáticos/fisiopatología , Glucagón/metabolismo , Hiperglucemia/etiología , Islotes Pancreáticos/metabolismo , Transmisión Sináptica , Animales , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Estimulación Eléctrica , Ganglios Simpáticos/efectos de los fármacos , Ganglios Simpáticos/metabolismo , Estimulantes Ganglionares/farmacología , Glucagón/sangre , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inervación , Masculino , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Agonistas Nicotínicos/farmacología , Norepinefrina/sangre , Norepinefrina/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Ratas Transgénicas , Ratas Wistar , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Transmisión Sináptica/efectos de los fármacos , Degeneración Walleriana/complicaciones
5.
bioRxiv ; 2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38854021

RESUMEN

Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (T IBAT , a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase T IBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9±2.0, 77.4±12.7 and 93.6±4.6% ( P <0.05), respectively and was unchanged in inguinal white adipose tissue, pancreas or liver. We subsequently measured the effects of acute 4V OT (1, 5 µg ≈ 0.99, 4.96 nmol) on T IBAT in DIO mice following sham or bilateral surgical SNS denervation to IBAT. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) elevated T IBAT similarly in sham mice as in denervated mice. We subsequently measured the effects of chronic 4V OT (16 nmol/day over 29 days) or vehicle infusions on body weight, adiposity and food intake in DIO mice following sham or bilateral surgical denervation of IBAT. Chronic 4V OT reduced body weight by 5.7±2.23% and 6.6±1.4% in sham and denervated mice ( P <0.05), respectively, and this effect was similar between groups ( P =NS). OT produced corresponding reductions in whole body fat mass ( P <0.05). Together, these findings support the hypothesis that sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and reductions of body weight and adiposity in male DIO mice.

6.
Front Endocrinol (Lausanne) ; 15: 1440070, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39145314

RESUMEN

Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (TIBAT, a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase TIBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9 ± 2.0, 77.4 ± 12.7 and 93.6 ± 4.6% (P<0.05), respectively and was unchanged in inguinal white adipose tissue, pancreas or liver. We subsequently measured the effects of acute 4V OT (1, 5 µg ≈ 0.99, 4.96 nmol) on TIBAT in DIO mice following sham or bilateral surgical SNS denervation to IBAT. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) elevated TIBAT similarly in sham mice as in denervated mice. We subsequently measured the effects of chronic 4V OT (16 nmol/day over 29 days) or vehicle infusions on body weight, adiposity and food intake in DIO mice following sham or bilateral surgical denervation of IBAT. Chronic 4V OT reduced body weight by 5.7 ± 2.23% and 6.6 ± 1.4% in sham and denervated mice (P<0.05), respectively, and this effect was similar between groups (P=NS). OT produced corresponding reductions in whole body fat mass (P<0.05). Together, these findings support the hypothesis that sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and reductions of body weight and adiposity in male DIO mice.


Asunto(s)
Tejido Adiposo Pardo , Adiposidad , Dieta Alta en Grasa , Ratones Endogámicos C57BL , Obesidad , Oxitocina , Sistema Nervioso Simpático , Animales , Oxitocina/farmacología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/inervación , Masculino , Ratones , Obesidad/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Adiposidad/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos , Ratones Obesos , Metabolismo Energético/efectos de los fármacos , Norepinefrina/metabolismo
8.
Diabetes ; 56(1): 217-23, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17192485

RESUMEN

Recurrent hypoglycemia impairs hormonal counterregulatory responses (CRRs) to further bouts of hypoglycemia. The hypothalamus and hindbrain are both critical for sensing hypoglycemia and triggering CRRs. Hypothalamic glucose sensing sites are implicated in the pathogenesis of defective CRRs; however, the contribution of hindbrain glucose sensing has not been elucidated. Using a rat model, we compared the effect of antecedent glucoprivation targeting hindbrain or hypothalamic glucose sensing sites with the effect of antecedent recurrent hypoglycemia on CRR to hypoglycemia induced 24 h later. Recurrent hypoglycemia decreased sympathoadrenal (1,470 +/- 325 vs. 3,811 +/- 540 pg/ml in controls [t = 60 min], P = 0.001) and glucagon secretion (222 +/- 43 vs. 494 +/- 56 pg/ml in controls [t = 60]), P = 0.003) in response to hypoglycemia. Antecedent 5-thio-glucose (5TG) injected into the hindbrain did not impair sympathoadrenal (3,806 +/- 344 pg/ml [t = 60]) or glucagon (513 +/- 56 pg/ml [t = 60]) responses to subsequent hypoglycemia. However, antecedent 5TG delivered into the third ventricle was sufficient to blunt CRRs to hypoglycemia. These results show that hindbrain glucose sensing is not involved in the development of defective CRRs. However, neural substrates surrounding the third ventricle are particularly sensitive to glucoprivic stimulation and may contribute importantly to the development of defective CRRs.


Asunto(s)
Glucosa/deficiencia , Glucosa/metabolismo , Hipoglucemia/fisiopatología , Rombencéfalo/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Glucemia/metabolismo , Ventrículos Cerebrales/metabolismo , Corticosterona/sangre , Epinefrina/sangre , Glucagón/sangre , Homeostasis , Masculino , Norepinefrina/sangre , Ratas , Ratas Sprague-Dawley , Recurrencia
9.
J Neurosci Res ; 86(9): 1981-93, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18338798

RESUMEN

Because damage to sympathetic nerve terminals occurs in a variety of diseases, we tested the hypothesis that nerve terminal damage per se is sufficient to impair ganglionic neurotransmission in vivo. First, we measured the effect of nerve terminal damage produced by the sympathetic nerve terminal toxin 6-hydroxydopamine (6-OHDA) on ganglionic levels of several neurotrophins thought to promote neurotransmission. 6-OHDA-induced nerve terminal damage did not decrease the expression of neurotrophin-4 or brain-derived neurotrophic factor mRNA in the celiac ganglia but did decrease the ganglionic content of both nerve growth factor protein (nadir = -63%) and the mRNA of the alpha-3 subunit of the nicotinic cholinergic receptor (nadir = -49%), a subunit required for neurotransmission. Next, we tested whether this degree of receptor deficiency was sufficient to impair activation of celiac ganglia neurons. Impaired fos mRNA responses to nicotine administration in the celiac ganglia of 6-OHDA-pretreated rats correlated temporally with suppressed expression of functional nicotinic receptors. We verified by Fos protein immunohistochemistry that this ganglionic impairment was specific to principal ganglionic neurons. Last, we tested whether centrally initiated ganglionic neurotransmission is also impaired following nerve terminal damage. The principal neurons in rat celiac ganglia were reflexively activated by 2-deoxy-glucose-induced glucopenia, and the Fos response in the celiac ganglia was markedly inhibited by pretreatment with 6-OHDA. We conclude that sympathetic nerve terminal damage per se is sufficient to impair ganglionic neurotransmission in vivo and that decreased nicotinic receptor production is a likely mediator.


Asunto(s)
Ganglios Simpáticos/fisiopatología , Terminaciones Nerviosas/patología , Sistema Nervioso Simpático/patología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Ganglios Simpáticos/efectos de los fármacos , Masculino , Factores de Crecimiento Nervioso/sangre , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Oxidopamina/toxicidad , ARN Mensajero/genética , Ratas , Ratas Wistar , Sistema Nervioso Simpático/efectos de los fármacos , Transmisión Sináptica
10.
Brain Res ; 1194: 65-72, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18191818

RESUMEN

The posterior paraventricular nucleus of the thalamus (THPVP) has been identified as a forebrain region that modulates the central nervous system (CNS) response to recurrent experiences of stressors. The THPVP is activated in response to a single (SH) or recurrent (RH) experience of the metabolic stress of hypoglycemia. In this study, we evaluated whether temporary experimental inactivation of the THPVP would modify the neuroendocrine response to SH or RH. Infusion of lidocaine (LIDO) or vehicle had no effect on the neuroendocrine response to SH, comparable to findings with other stressors. THPVP vehicle infusion concomitant with RH resulted in a prevention of the expected impairment of neuroendocrine responses, relative to SH. LIDO infusion with RH resulted in significantly decreased glucagon and sympathoadrenal responses, relative to SH. These results suggest that the THPVP may contribute to the sympathoadrenal stimulation induced by hypoglycemia; and emphasizes that the THPVP is a forebrain region that may contribute to the coordinated CNS response to metabolic stressors.


Asunto(s)
Glucagón/metabolismo , Hipoglucemia/fisiopatología , Núcleos Talámicos de la Línea Media/fisiología , Sistemas Neurosecretores/fisiología , Corticoesteroides/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Análisis de Varianza , Anestésicos Locales/farmacología , Animales , Glucemia , Epinefrina/metabolismo , Hipoglucemia/inducido químicamente , Insulina , Lidocaína/farmacología , Masculino , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Sistemas Neurosecretores/efectos de los fármacos , Norepinefrina/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
11.
J Clin Endocrinol Metab ; 92(10): 4035-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17684049

RESUMEN

CONTEXT: Older studies have shown that high doses of norepinephrine infused into human subjects can inhibit insulin secretion. Similar inhibition during electrical stimulation of sympathetic nerves in animals raises the possibility that the suppression of insulin secretion seen in humans could reflect a physiological effect of sympathetic nerves on islet beta-cells. However, a direct test of the hypothesis that moderate and selective activation of these nerves is sufficient to inhibit insulin secretion in humans is lacking. OBJECTIVE: We sought to test this hypothesis by releasing moderate amounts of endogenous norepinephrine selectively from the sympathetic nerves of normal human subjects by infusing them with low doses of the indirect sympathomimetic agent tyramine. METHODS: During a single study visit, 11 healthy subjects received iv injections of arginine either alone or in combination with a low-dose tyramine infusion. Physiological (blood pressure) and biochemical (insulin, glucose, and norepinephrine) parameters were measured. RESULTS: The acute insulin response to arginine was significantly reduced during tyramine compared with that seen in the absence of tyramine (P = 0.036). CONCLUSIONS: These data suggest that moderate and selective activation of sympathetic nerves inhibits insulin release in humans.


Asunto(s)
Insulina/metabolismo , Sistema Nervioso Simpático/fisiología , Simpatomiméticos/administración & dosificación , Tiramina/administración & dosificación , Adolescente , Adulto , Arginina/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Femenino , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Norepinefrina/sangre , Sistema Nervioso Simpático/efectos de los fármacos
12.
Diabetes ; 66(4): 823-834, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28115396

RESUMEN

Dynamic adjustment of insulin secretion to compensate for changes of insulin sensitivity that result from alteration of nutritional or metabolic status is a fundamental aspect of glucose homeostasis. To investigate the role of the brain in this coupling process, we used cold exposure as an experimental paradigm because the sympathetic nervous system (SNS) helps to coordinate the major shifts of tissue glucose utilization needed to ensure that increased thermogenic needs are met. We found that glucose-induced insulin secretion declined by 50% in rats housed at 5°C for 28 h, and yet, glucose tolerance did not change, owing to a doubling of insulin sensitivity. These potent effects on insulin secretion and sensitivity were fully reversed by returning animals to room temperature (22°C) for 4 h or by intravenous infusion of the α-adrenergic receptor antagonist phentolamine for only 30 min. By comparison, insulin clearance was not affected by cold exposure or phentolamine infusion. These findings offer direct evidence of a key role for the brain, acting via the SNS, in the rapid, highly coordinated, and reciprocal changes of insulin secretion and insulin sensitivity that preserve glucose homeostasis in the setting of cold exposure.


Asunto(s)
Glucemia/metabolismo , Frío , Resistencia a la Insulina , Insulina/metabolismo , Sistema Nervioso Simpático/metabolismo , Antagonistas Adrenérgicos alfa/farmacología , Animales , Glucemia/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Secreción de Insulina , Masculino , Fentolamina/farmacología , Ratas , Ratas Long-Evans , Ratas Wistar , Sistema Nervioso Simpático/efectos de los fármacos
13.
Endocrinology ; 147(6): 2893-901, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16527847

RESUMEN

The hormone ghrelin is secreted mainly from the gut, rises in peripheral plasma before meals, and is implicated in stimulating hunger, initiating meals, and developing obesity. We hypothesize that activation of the sympathetic nervous system contributes to preprandial ghrelin surges. The present studies in isoflurane-anesthetized Wistar rats were designed to determine whether sympathetic nerves and neurohormones are capable of stimulating ghrelin secretion. We activated gut sympathetic nerves by two methods: electrical sympathetic nerve stimulation (SNS) and chemical sympathetic nerve activation with iv tyramine (TYR) administration. Portal venous blood was sampled before and during a 10-min sympathetic stimulation. Successful activation of gut sympathetic nerves was verified by increments in portal venous norepinephrine. SNS increased portal ghrelin by 206 +/- 50%. In contrast, simply isolating gut sympathetic nerves without applying current had a minimal effect on ghrelin levels. TYR also increased portal ghrelin [change (Delta), +52 +/- 11%], whereas saline infusion had little effect. We next determined whether the neural stimulation of ghrelin secretion was mediated indirectly via the suppression of insulin secretion during SNS and TYR. Streptozotocin-induced diabetes prevented a fall in insulin during TYR, yet the portal ghrelin response (Delta = +47 +/- 18%) was similar to that in nondiabetic rats. Lastly, to test for humoral stimulation of ghrelin, we infused the sympathetic neurohormone, epinephrine, to achieve levels found during severe stress. Epinephrine failed to stimulate ghrelin secretion (Delta = +4 +/- 35%). We conclude that the neural, but not the neurohumoral, branch of the sympathetic nervous system can directly stimulate ghrelin secretion.


Asunto(s)
Hormonas Peptídicas/metabolismo , Sistema Nervioso Simpático/fisiología , Animales , Estimulación Eléctrica , Epinefrina/farmacología , Ghrelina , Insulina/metabolismo , Secreción de Insulina , Masculino , Norepinefrina/metabolismo , Ratas , Ratas Wistar , Estreptozocina/farmacología , Tirosina/farmacología
14.
Neuropeptides ; 40(1): 1-10, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16487586

RESUMEN

BB rats lose >50% of their islet sympathetic nerve terminals soon after diabetes onset, markedly impairing the glucagon response to activation of these nerves. In this study, we sought evidence that this degree of disease-induced nerve terminal damage affected their neuronal cell bodies. Increased galanin expression was used as a marker of the change of phenotype that occurs in neuronal cell bodies when their axons are severely damaged. The celiac ganglion (CG) was analyzed because it is a major source of the sympathetic nerves that project to the pancreatic islets. But we first needed to determine if damaging nerve terminals could increase galanin expression in this ganglion and, if so, when that expression was maximal. Severe, global nerve terminal damage produced a dramatic increase of CG galanin expression which was maximal 5 days later. We next determined if a global, but partial, nerve terminal loss would also increase galanin expression and found a significant increase of galanin mRNA and its peptide in the CG. Finally, we determined if the disease-induced, partial and islet-selective loss of nerve terminals seen in BB diabetic rats was sufficient to increase galanin: we, again, found a significant increase of galanin mRNA and its peptide in their CG. These increases did not occur in their superior cervical ganglia. We conclude that the selective damage to islet sympathetic nerve terminals seen in BB diabetic rats, rather than the systemic factors of diabetic hyperglycemia or insulin deficiency, causes the increased galanin expression observed in the CG of this animal model of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Galanina/genética , Ganglios Simpáticos/fisiopatología , Animales , Modelos Animales de Enfermedad , Ganglios Simpáticos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Oxidopamina/farmacología , ARN Mensajero/genética , Ratas , Ratas Endogámicas BB , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Diabetes ; 65(8): 2322-30, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27207540

RESUMEN

In humans, the glucagon response to moderate-to-marked insulin-induced hypoglycemia (IIH) is largely mediated by the autonomic nervous system. Because this glucagon response is impaired early in type 1 diabetes, we sought to determine if these patients, like animal models of autoimmune diabetes, have an early and severe loss of islet sympathetic nerves. We also tested whether this nerve loss is a permanent feature of type 1 diabetes, is islet-selective, and is not seen in type 2 diabetes. To do so, we quantified pancreatic islet and exocrine sympathetic nerve fiber area from autopsy samples of patients with type 1 or 2 diabetes and control subjects without diabetes. Our central finding is that patients with either very recent onset (<2 weeks) or long duration (>10 years) of type 1 diabetes have a severe loss of islet sympathetic nerves (Δ = -88% and Δ = -79%, respectively). In contrast, patients with type 2 diabetes lose no islet sympathetic nerves. There is no loss of exocrine sympathetic nerves in either type 1 or type 2 diabetes. We conclude that patients with type 1, but not type 2, diabetes have an early, marked, sustained, and islet-selective loss of sympathetic nerves, one that may impair their glucagon response to IIH.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Sistema Nervioso Simpático/patología , Adolescente , Adulto , Sistema Nervioso Autónomo/metabolismo , Sistema Nervioso Autónomo/patología , Sistema Nervioso Autónomo/fisiopatología , Niño , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Glucagón/metabolismo , Humanos , Hipoglucemia/metabolismo , Hipoglucemia/patología , Hipoglucemia/fisiopatología , Inmunohistoquímica , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Islotes Pancreáticos/fisiopatología , Masculino , Persona de Mediana Edad , Páncreas/metabolismo , Páncreas/patología , Páncreas/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Adulto Joven
16.
Diabetes ; 51(10): 2997-3002, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12351439

RESUMEN

To discover whether islet sympathetic nerves are damaged during the autoimmune destruction of islet B-cells, we immunostained sections of pancreas from BioBreeder (BB) diabetic rats, using antibodies against vesicular monoamine transporter 2 (VMAT2), a marker of sympathetic nerve terminals. We found a marked decrease in the VMAT2-positive fiber area in the islets of BB rats that had been diabetic for only 1-2 weeks compared with their nondiabetic controls. In contrast, there was no significant decrease in the VMAT2-positive fiber area in the exocrine pancreas in these early diabetic BB rats. Furthermore, streptozotocin-diabetic rats showed no decrease in VMAT2-positive fiber area in their islets compared with controls. The classical diabetic autonomic neuropathy (DAN) that eventually occurs in the heart was not present in BB diabetic rats at this early stage as evidenced by normal cardiac VMAT2 immunostaining and normal cardiac norepinephrine content. Also, in contrast to DAN, this islet neuropathy did not worsen with duration of diabetes. These data provide evidence of a heretofore unrecognized early sympathetic islet neuropathy (eSIN). Because eSIN occurs selectively in the islet, is rapid in onset, and is associated with autoimmune but not chemically induced diabetes, it is distinct from DAN in location, time course, and mechanism.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Islotes Pancreáticos/inervación , Islotes Pancreáticos/patología , Proteínas de Transporte de Membrana , Neuropéptidos , Sistema Nervioso Simpático/patología , Animales , Anticuerpos , Diabetes Mellitus Tipo 1/genética , Femenino , Masculino , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/inmunología , Fibras Nerviosas/química , Fibras Nerviosas/patología , Oxidopamina , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Ratas , Ratas Endogámicas BB , Simpatectomía Química , Simpaticolíticos , Proteínas de Transporte Vesicular de Aminas Biógenas , Proteínas de Transporte Vesicular de Monoaminas
18.
Diabetes ; 64(7): 2376-87, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25633417

RESUMEN

Several lines of evidence implicate excess glucagon secretion in the elevated rates of hepatic glucose production (HGP), hyperglycemia, and ketosis characteristic of uncontrolled insulin-deficient diabetes (uDM), but whether hyperglucagonemia is required for hyperglycemia in this setting is unknown. To address this question, adult male Wistar rats received either streptozotocin (STZ) to induce uDM (STZ-DM) or vehicle and remained nondiabetic. Four days later, animals received daily subcutaneous injections of either the synthetic GLP-1 receptor agonist liraglutide in a dose-escalating regimen to reverse hyperglucagonemia or its vehicle for 10 days. As expected, plasma glucagon levels were elevated in STZ-DM rats, and although liraglutide treatment lowered glucagon levels to those of nondiabetic controls, it failed to attenuate diabetic hyperglycemia, elevated rates of glucose appearance (Ra), or increased hepatic gluconeogenic gene expression. In contrast, it markedly reduced levels of both plasma ketone bodies and hepatic expression of the rate-limiting enzyme involved in ketone body production. To independently confirm this finding, in a separate study, treatment of STZ-DM rats with a glucagon-neutralizing antibody was sufficient to potently lower plasma ketone bodies but failed to normalize elevated levels of either blood glucose or Ra. These data suggest that in rats with uDM, hyperglucagonemia is required for ketosis but not for increased HGP or hyperglycemia.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Glucagón/sangre , Glucosa/metabolismo , Hiperglucemia/etiología , Cetosis/etiología , Hígado/metabolismo , Animales , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/fisiología , Glucagón/fisiología , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/farmacología , Hiperglucemia/sangre , Insulina/farmacología , Cuerpos Cetónicos/sangre , Cetosis/sangre , Liraglutida , Masculino , Ratas Wistar , Receptores de Glucagón/fisiología , Estreptozocina
19.
Mol Metab ; 4(8): 561-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26266088

RESUMEN

OBJECTIVE: Central administration of ligands for fibroblast growth factor receptors (FGFRs) such as fibroblast growth factor-19 (FGF19) and FGF21 exert glucose-lowering effects in rodent models of obesity and type 2 diabetes (T2D). Conversely, intracerebroventricular (icv) administration of the non-selective FGFR inhibitor (FGFRi) PD173074 causes glucose intolerance, implying a physiological role for neuronal FGFR signaling in glucose homeostasis. The current studies were undertaken to identify neuroendocrine mechanisms underlying the glucose intolerance induced by pharmacological blockade of central FGFRs. METHODS: Overnight fasted, lean, male, Long-Evans rats received icv injections of either PD173074 or vehicle (Veh) followed 30 min later by performance of a frequently sampled intravenous glucose tolerance test (FSIGT). Minimal model analysis of glucose and insulin data from the FSIGT was performed to estimate insulin-dependent and insulin-independent components of glucose disposal. Plasma levels of lactate, glucagon, corticosterone, non-esterified free fatty acids (NEFA) and catecholamines were measured before and after intravenous (iv) glucose injection. RESULTS: Within 20 min of icv PD173074 injection (prior to the FSIGT), plasma levels of lactate, norepinephrine and epinephrine increased markedly, and each returned to baseline rapidly (within 8 min) following the iv glucose bolus. In contrast, plasma glucagon levels were not altered by icv FGFRi at either time point. Consistent with a previous report, glucose tolerance was impaired following icv PD173074 compared to Veh injection and, based on minimal model analysis of FSIGT data, this effect was attributable to reductions of both insulin secretion and the basal insulin effect (BIE), consistent with the inhibitory effect of catecholamines on pancreatic ß-cell secretion. By comparison, there were no changes in glucose effectiveness at zero insulin (GEZI) or the insulin sensitivity index (SI). To determine if iv glucose (given during the FSIGT) contributed to the rapid resolution of the sympathoadrenal response induced by icv FGFRi, we performed an additional study comparing groups that received iv saline or iv glucose 30 min after icv FGFRi. Our finding that elevated plasma catecholamine levels returned rapidly to baseline irrespective of whether rats subsequently received an iv bolus of saline or glucose indicates that the rapid reversal of sympathoadrenal activation following icv FGFRi was unrelated to the subsequent glucose bolus. CONCLUSIONS: The effect of acute inhibition of central FGFR signaling to impair glucose tolerance likely involves a stress response associated with pronounced, but transient, sympathoadrenal activation and an associated reduction of insulin secretion. Whether this effect is a true consequence of FGFR blockade or involves an off-target effect of the FGFR inhibitor requires additional study.

20.
Metabolism ; 51(1): 12-9, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11782866

RESUMEN

To gain insight into the mechanisms responsible for the loss of the glucagon response to insulin-induced hypoglycemia in type 1 diabetes, glucagon responses to 4 different stimuli were examined over 3 months of diabetes in alloxan-treated mice. At 1, 6, and 12 weeks after alloxan (60 mg/kg), phloridzin (0.1 g/kg) was administered to overnight fasted diabetic mice to match the glucose levels of those in nondiabetic control mice before administration of the acute stimuli. Despite the elevation of baseline glucagon levels produced by the phloridzin treatment, the glucagon responses to insulin (2 U/kg intraperitoneally [IP])-induced hypoglycemia was not impaired at 1 week. However, the response was reduced by greater than 60% after 6 and 12 weeks of diabetes (P <.05). In contrast, the glucagon response to arginine (0.25 g/kg intravenously [IV]) was not reduced after 1, 6, or 12 weeks of diabetes, ruling out a generalized impairment of the A-cell responses. The glucagon response to the neuroglucopenic agent, 2-deoxyglucose (2-DG; 500 mg/kg IV) was impaired, like that to insulin-induced hypoglycemia, after 6 and 12 weeks of diabetes (P <.05), suggesting that supersensitivity to the potential inhibitory effects of exogenous insulin is not the mechanism responsible for the impairment. Finally, the glucagon response to the cholinergic agonist, carbachol (0.53 micromol/kg IV), was not impaired in the diabetic animals, arguing against a defect in the A-cell's responsiveness to autonomic stimulation. The data suggest that the impairment of the glucagon response to insulin-induced hypoglycemia in alloxan diabetic mice is not due to a generalized impairment of A-cell responsiveness, to desensitization by a suppressive action of insulin, or to impairment of the A-cell response to autonomic stimuli. The remaining mechanisms which are likely to explain the late loss of the glucagon response to insulin-induced hypoglycemia include (1) a defect in the A-cell recognition of glucopenic stimuli, or (2) a defect in the autonomic inputs to the A cell that are known to be activated by glucopenic stimuli.


Asunto(s)
Arginina/farmacología , Encéfalo/metabolismo , Carbacol/farmacología , Diabetes Mellitus Experimental/sangre , Glucagón/sangre , Glucosa/deficiencia , Hipoglucemia/sangre , Animales , Desoxiglucosa/farmacología , Femenino , Hipoglucemia/inducido químicamente , Hipoglucemiantes , Insulina , Ratones , Ratones Endogámicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA