Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters











Publication year range
1.
J Neuroendocrinol ; 30(1)2018 01.
Article in English | MEDLINE | ID: mdl-29224246

ABSTRACT

Among vertebrate species of the major vertebrate classes in the wild, a seasonal rhythm of whole body fuel metabolism, oscillating from a lean to obese condition, is a common biological phenomenon. This annual cycle is driven in part by annual changes in the circadian dopaminergic signalling at the suprachiasmatic nuclei (SCN), with diminution of circadian peak dopaminergic activity at the SCN facilitating development of the seasonal obese insulin-resistant condition. The present study investigated whether such an ancient circadian dopamine-SCN activity system for expression of the seasonal obese, insulin-resistant phenotype may be operative in animals made obese amd insulin resistant by high-fat feeding and, if so, whether reinstatement of the circadian dopaminergic peak at the SCN would be sufficient to reverse the adverse metabolic impact of the high-fat diet without any alteration of caloric intake. First, we identified the supramammillary nucleus as a novel site providing the majority of dopaminergic neuronal input to the SCN. We further identified dopamine D2 receptors within the peri-SCN region as being functional in mediating SCN responsiveness to local dopamine. In lean, insulin-sensitive rats, the peak in the circadian rhythm of dopamine release at the peri-SCN coincided with the daily peak in SCN electrophysiological responsiveness to local dopamine administration. However, in rats made obese and insulin resistant by high-fat diet (HFD) feeding, these coincident circadian peak activities were both markedly attenuated or abolished. Reinstatement of the circadian peak in dopamine level at the peri-SCN by its appropriate circadian-timed daily microinjection to this area (but not outside this circadian time-interval) abrogated the obese, insulin-resistant condition without altering the consumption of the HFD. These findings suggest that the circadian peak of dopaminergic activity at the peri-SCN/SCN is a key modulator of metabolism and the responsiveness to adverse metabolic consequences of HFD consumption.


Subject(s)
Circadian Rhythm/physiology , Diet, High-Fat , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Biological Clocks , Female , Hypothalamus, Posterior/metabolism , Obesity/metabolism , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism
2.
Metabolism ; 50(11): 1377-84, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11699060

ABSTRACT

Sympatholytic dopamine agonist treatment utilizing bromocriptine and SKF38393 (BC/SKF) significantly lowers basal plasma insulin levels and normalizes basal and glucose-induced insulin secretion of the pancreatic beta cell in ob/ob mice. While BC/SKF has no significant effect on pancreatic islet cells directly, drug action is mediated via alterations in the hypothalamic-neuroendocrine axis, which drives metabolic changes in peripheral tissues leading to a marked reduction in hyperglycemia and hyperlipidemia and corrects autonomic control of islet function. To elucidate the nature of the functional response of islets to systemic BC/SKF treatment in ob/ob mice, we investigated the relative changes in the levels of functionally important beta-cell proteins in situ, as well as differences in the beta-cell turnover rate, following a 2-week drug treatment. Isolated islets from treated mice exhibit a 3.5-fold increase in insulin content (P <.01) that correlated with a 51% reduction in basal plasma insulin levels (P <.01) compared with vehicle-treated controls. Using quantitative immunofluorescence microscopy on pancreatic tissue sections, insulin and GLUT2 immunoreactivity of islet beta cells of BC/SKF-treated mice were significantly increased (approximately 2.3-fold and approximately 4.4-fold, respectively; P <.002) to the levels observed in islets of their lean littermates. Glucokinase (GK) immunoreactivity was greatly (75%) reduced in beta cells from ob/ob versus lean mice (P <.0001). A modest increase in GK immunoreactivity in beta cells of drug-treated mice was observed (approximately 1.6-fold; P <.05). Isolated islets from BC/SKF-treated mice exhibit a 42% reduction in DNA content compared with vehicle-treated controls (P <.01) to levels observed in lean mice, but without notable differences in islet size. In situ assays for mitosis and apoptosis, using 5-bromodeoxyuridine (BrdU) and terminal deoxyribotransferase (TdT)-UTP nick end labeling (TUNEL) staining techniques, respectively, were performed in pancreas of these mice to determine if beta cells show a reduction in hyperplasia following BC/SKF treatment. Accordingly, a pronounced decrease in replicating, BrdU-positive beta cells in the drug-treated mice compared with the control group was observed, but without differences in their TUNEL-staining patterns. Collectively, these data suggest that systemic sympatholytic dopaminergic therapy that attenuates hyperglycemia and hyperlipidemia improves islet function in ob/ob mice by improving aberrations in the beta cell's glucose-sensing apparatus, enhancing insulin storage and/or retention, and stabilizing hyperplasia, thus reducing basal insulin levels.


Subject(s)
Dopamine Agonists/pharmacology , Glucokinase/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Monosaccharide Transport Proteins/metabolism , Sympatholytics/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , DNA/metabolism , Female , Glucagon/metabolism , Glucose Transporter Type 2 , Hyperplasia/pathology , Hyperplasia/prevention & control , Immunohistochemistry , In Situ Nick-End Labeling , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Inbred C57BL , Mice, Obese , Somatostatin/metabolism
3.
Int J Obes Relat Metab Disord ; 25(5): 698-704, 2001 May.
Article in English | MEDLINE | ID: mdl-11360153

ABSTRACT

OBJECTIVE: Several studies have implicated increased sympathetic tone as a contributing factor to the hyperglycemia and hyperglucagonemia of ob/ob mice. However, the responsiveness of plasma glucose, insulin and glucagon to circulating norepinephrine (NE) in ob/ob vs normal lean mice has never been described. Therefore, the present study investigated the effect of a 15 min intravenous NE infusion (1 pmol/min/g) on plasma glucose, insulin and glucagon in anesthetized lean, ob/ob, ob/ob-concurrent yohimbine (alpha(2) antagonist) treated, and ob/ob-chronically sympatholytic dopamine agonist treated (for 14 days prior to infusion) mice. In an effort to gain insight into a possible relation between norepinephrine, hyperglucagonemia and hyperinsulinemia in ob/ob mice, this study also examined the isolated islet responses to NE and glucagon in lean, ob/ob and ob/ob-sympatholytic dopamine agonist treated mice. RESULTS: Basal humoral values of glucose, insulin and glucagon were all elevated in ob/ob vs lean mice (by 63, 1900 and 63%, respectively, P<0.01). However, NE infusion further increased levels of glucose, insulin and glucagon in ob/ob (by 80, 90 and 60%, respectively, P<0.05) but not in lean mice (between group difference for all parameters P<0.05). Acute concurrent yohimbine treatment as well as chronic prior sympatholytic dopamine agonist treatment (bromocriptine plus SKF38393) simultaneously strongly aborgated or abolished all these humoral hypersensitivity responses to intravenous NE in ob/ob mice (P<0.05). Clamping the plasma glucose level in untreated ob/ob mice at a high level (30 mM) established by NE infusion did not significantly alter the plasma insulin level, suggesting that some other influence of NE was responsible for this insulin effect. Direct NE administration at 1 microM to islets from lean and ob/ob mice inhibited 15 mM glucose-stimulated insulin secretion in both groups, but at 0.1 microM it was inhibitory only in islets from ob/ob mice. However, glucagon (10 nM) increased 15 mM glucose-stimulated insulin secretion in ob/ob (by 170%, P<0.05) but not lean mice (between group difference P<0.05). CONCLUSION: These findings suggest that hypersensitivity to circulating NE may potentiate hyperglycemia and hyperglucagonemia in ob/ob mice, and the subsequent hyperglucagonemia coupled with increased islet beta-cell insulin secretory responsiveness to glucagon in ob/ob mice may support hyperinsulinemia, thus explaining the increased plasma insulin level response to intravenous NE in these animals. These findings further support a role for increased peripheral noradrenergic activities in the development and maintenance of the hyperglycemic, hyperglucagonemic and hyperinsulinemic state, characteristic of type 2 diabetes.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Blood Glucose/metabolism , Glucagon/metabolism , Insulin/metabolism , Norepinephrine/pharmacology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Adrenergic alpha-Agonists/administration & dosage , Animals , Bromocriptine/pharmacology , Dopamine Agonists/pharmacology , Female , Glucose Clamp Technique , Hyperglycemia/etiology , Hyperinsulinism/etiology , Insulin Secretion , Mice , Mice, Inbred C57BL , Mice, Obese , Norepinephrine/administration & dosage , Norepinephrine/physiology , Random Allocation
4.
Diabetes Care ; 23(8): 1154-61, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10937514

ABSTRACT

OBJECTIVE: In vertebrates, body fat stores and insulin action are controlled by the temporal interaction of circadian neuroendocrine oscillations. Bromocriptine modulates neurotransmitter action in the brain and has been shown to improve glucose tolerance and insulin resistance in animal models of obesity and diabetes. We studied the effect of a quick-release bromocriptine formulation on glucose homeostasis and insulin sensitivity in obese type 2 diabetic subjects. RESEARCH DESIGN AND METHODS: There were 22 obese subjects with type 2 diabetes randomized to receive a quick-release formulation of bromocriptine (n = 15) or placebo (n = 7) in a 16-week double-blind study. Subjects were prescribed a weight-maintaining diet to exclude any effect of changes in body weight on the primary outcome measurements. Fasting plasma glucose concentration and HbA(1c) were measured at 2- to 4-week intervals during treatment. Body composition (underwater weighing), body fat distribution (magnetic resonance imaging), oral glucose tolerance (oral glucose tolerance test [OGTT]), insulin-mediated glucose disposal, and endogenous glucose production (2-step euglycemic insulin clamp, 40 and 160 mU x min(-1) x m(-2)) were measured before and after treatment. RESULTS: No changes in body weight or body composition occurred during the study in either placebo- or bromocriptine-treated subjects. Bromocriptine significantly reduced HbA(1c) (from 8.7 to 8.1%, P = 0.009) and fasting plasma glucose (from 190 to 172 mg/dl, P = 0.02) levels, whereas these variables increased during placebo treatment (from 8.5 to 9.1%, NS, and from 187 to 223 mg/dl, P = 0.02, respectively). The differences in HbA(1c) (delta = 1.2%, P = 0.01) and fasting glucose (delta = 54 mg/dl, P < 0.001) levels between the bromocriptine and placebo group at 16 weeks were highly significant. The mean plasma glucose concentration during OGTT was significantly reduced by bromocriptine (from 294 to 272 mg/dl, P = 0.005), whereas it increased in the placebo group. No change in glucose disposal occurred during the first step of the insulin clamp in either the bromocriptine- or placebo-treated group. During the second insulin clamp step, bromocriptine improved total glucose disposal from 6.8 to 8.4 mg x min(-1) kg(-1) fat-free mass (FFM) (P = 0.01) and nonoxidative glucose disposal from 3.3 to 4.3 mg min(-1) x kg(-1) FFM (P < 0.05), whereas both of these variables deteriorated significantly (P < or = 0.02) in the placebo group. CONCLUSIONS: Bromocriptine improves glycemic control and glucose tolerance in obese type 2 diabetic patients. Both reductions in fasting and postprandial plasma glucose levels appear to contribute to the improvement in glucose tolerance. The bromocriptine-induced improvement in glycemic control is associated with enhanced maximally stimulated insulin-mediated glucose disposal.


Subject(s)
Bromocriptine/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus/drug therapy , Obesity , Abdomen , Adipose Tissue/anatomy & histology , Blood Glucose/metabolism , Body Mass Index , Body Weight , Diabetes Mellitus/blood , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Diet, Diabetic , Double-Blind Method , Female , Glucose Tolerance Test , Glycated Hemoglobin/analysis , Hormone Antagonists/therapeutic use , Humans , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Placebos , Time Factors , Viscera
5.
Am J Physiol Regul Integr Comp Physiol ; 279(2): R505-14, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10938239

ABSTRACT

The genetically, seasonally, and diet-induced obese, glucose-intolerant states in rodents, including ob/ob mice, have each been associated with elevated hypothalamic levels of norepinephrine (NE). With the use of quantitative autoradiography on brain slices of 6-wk-old obese (ob/ob) and lean mice, the adrenergic receptor populations in several hypothalamic nuclei were examined. The binding of [(125)I]iodocyanopindolol to beta(1)- and beta(2)-adrenergic receptors in ob/ob mice was significantly increased in the paraventricular hypothalamic nucleus (PVN) by 30 and 38%, in the ventromedial hypothalamus (VMH) by 23 and 72%, and in the lateral hypothalamus (LH) by 10 and 15%, respectively, relative to lean controls. The binding of [(125)I]iodo-4-hydroxyphenyl-ethyl-aminomethyl-tetralone to alpha(1)-adrenergic receptors was also significantly increased in the PVN (26%), VMH (67%), and LH (21%) of ob/ob mice. In contrast, the binding of [(125)I]paraiodoclonidine to alpha(2)-adrenergic receptors in ob/ob mice was significantly decreased in the VMH (38%) and the dorsomedial hypothalamus (17%) relative to lean controls. This decrease was evident in the alpha(2A)- but not the alpha(2BC)-receptor subtype. Scatchard analysis confirmed this decreased density of alpha(2)-receptors in ob/ob mice. Together with earlier studies, these changes in hypothalamic adrenergic receptors support a role for increased hypothalamic NE activity in the development of the metabolic syndrome of ob/ob mice.


Subject(s)
Hypothalamus/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, beta/metabolism , Tetralones , Animals , Autoradiography , Clonidine/analogs & derivatives , Clonidine/metabolism , Female , In Vitro Techniques , Iodocyanopindolol/metabolism , Mice , Phenethylamines/metabolism , Reference Values , Tissue Distribution
6.
Chronobiol Int ; 17(2): 155-72, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10757461

ABSTRACT

Bromocriptine, a dopamine D2 agonist, inhibits seasonal fattening and improves seasonal insulin resistance in Syrian hamsters. Alterations in daily rhythms of neuroendocrine activities are involved in the regulation of seasonal metabolic changes. Changes in circadian neuroendocrine activities that regulate metabolism are believed to be modulated by central circadian oscillators within the hypothalamic suprachiasmatic nuclei (SCN) of seasonal animals. We examined the association of metabolic responses to bromocriptine with its effects on the daily rhythms of metabolic hormones and daily monoamine profiles within the SCN, a primary circadian pacemaker known to regulate metabolism, in Syrian hamsters. Obese glucose-intolerant male Syrian hamsters (body weight [BW] 185 +/- 10 g) held on 14h daily photoperiods were treated at light onset with bromocriptine (800 microg/animal/day, ip) or vehicle for 2 weeks. Animals were then subjected to a glucose tolerance test (GTT) (3 g/kg BW, ip). Different subsets of animals (n = 6) from each treatment group were sacrificed at 0h/24h, 5h, 10h, 15h, or 20h after light onset for analyses of SCN monoamines, plasma insulin, prolactin, cortisol, thyroxin (T4), triiodothyronine (T3), glucose, and free fatty acids (FFAs). Compared with control values, bromocriptine treatment significantly reduced weight gain (14.9 vs. -2.9 g, p < .01) and the areas under the GTT glucose and insulin curves by 29% and 48%, respectively (p < .05). Basal plasma insulin concentration was markedly reduced throughout the day in bromocriptine-treated animals without influencing plasma glucose levels. Bromocriptine reduced the daily peak in FFA by 26% during the late light span (p < .05). Bromocriptine significantly shifted the daily plasma cortisol peak from the early dark to the light period of the day, reduced the plasma prolactin (mean 1.8 vs. 39.4 ng/dL) and T4 throughout the day (mean 1.6 vs. 3.8 microg/dL), and selectively reduced T3 during the dark period of the day (p < .01). Concurrently, bromocriptine treatment significantly reduced SCN dopamine turnover during the light period and shifted daily peaks of SCN serotonin and 5-hydroxy-indoleacetic acid (5-HIAA) content by 12h from the light to the dark period of the day (p < .05). This was confirmed by a further in vivo microdialysis study in which bromocriptine increased SCN extracellular 5-HIAA of glucose-intolerant hamsters during the dark phase (47% increase, p < .05) toward levels observed in normal glucose-tolerant hamsters. Thus, bromocriptine-induced resetting of daily patterns of SCN neurotransmitter metabolism is associated with the effects of bromocriptine on attenuation of the obese insulin-resistant and glucose-intolerant condition. A large body of corroborating evidence suggests that such bromocriptine-induced changes in SCN monoamine metabolism may be functional in its effects on metabolism.


Subject(s)
Biogenic Monoamines/metabolism , Bromocriptine/pharmacology , Circadian Rhythm/drug effects , Dopamine Agonists/pharmacology , Hypoglycemic Agents/pharmacology , Suprachiasmatic Nucleus/metabolism , Animals , Blood Glucose/physiology , Body Weight/drug effects , Cricetinae , Dopamine/metabolism , Fatty Acids, Nonesterified/blood , Glucose Intolerance/metabolism , Glucose Tolerance Test , Hormones/blood , Insulin Resistance/physiology , Male , Mesocricetus , Obesity/metabolism , Serotonin/metabolism , Suprachiasmatic Nucleus/drug effects
7.
Int J Mol Med ; 5(4): 349-55, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10719049

ABSTRACT

Studies of the effects of acute and chronic norepinephrine (NE) infusion into the ventromedial hypothalamus (VMH) of rodents indicate important roles for VMH NE activities in the development of the obese-glucose intolerant state. Moreover, elevated endogenous levels of NE and/or its metabolites have been observed in a variety of obese-glucose intolerant animal models. We therefore investigated the VMH neuronal electrophysiologic responsiveness to iontophoretically applied NE in lean-euglycemic and obese-hyperglycemic mice. Additionally, the effect of dopamine agonist treatment (which reduces obesity and hyperglycemia) on VMH responsiveness to NE was examined in obese-hyperglycemic mice. Obese (ob/ob) mice were treated daily for 14 days with either bromocriptine (BC, D2 agonist) (10 mg/kg) plus SKF38393 (SKF, D1 agonist) (20 mg/kg) or vehicle. Lean mice were also similarly treated with vehicle. Twenty-seven hours following the final treatment, mice were anesthetized to obtain electrophysiologic responses of glutamate activated VMH neurons to local NE administration. In all three study groups, NE administration inhibited glutamate evoked neuronal activity in the majority (90%) of recorded neurons. No response to NE was observed in the remaining 10% of neurons. Also within all three groups there existed two patterns of response to NE; a) long duration (>2 min) and low threshold (<20 nA) and b) short duration and high threshold. Relative to lean mice, obese mice exhibited a significant 70% increase in average duration of response, 3-fold increase in percent neurons with long duration of response, and 2-fold increase in percent neurons with low threshold of response. BC/SKF treatment of obese mice significantly reduced the percent VMH neurons with long duration and low threshold of response to NE to resemble the VMH neuronal responsiveness to NE observed in lean mice. Increased VMH responsiveness to NE is part of the endogenous neurophysiology of obese-hyperglycemic ob/ob mice. Taken together with previous findings mentioned above, the present results suggest that this increased VMH responsiveness to NE contributes to the pathophysiology of the obese-hyperglycemic state.


Subject(s)
Hyperglycemia/physiopathology , Neurons/physiology , Norepinephrine/pharmacology , Obesity/physiopathology , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/physiopathology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Bromocriptine/pharmacology , Dopamine Agonists/pharmacology , Electrophysiology , Female , Glutamic Acid/pharmacology , Hyperglycemia/prevention & control , Iontophoresis , Mice , Mice, Inbred C57BL , Mice, Obese , Neurons/drug effects , Obesity/prevention & control , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Syndrome
8.
Neuroreport ; 11(2): 383-7, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10674491

ABSTRACT

Numerous studies have implicated increased ventromedial hypothalamic (VMH) norepinephrine (NE) activity as a contributing factor to the obese, hyperinsulinemic, glucose intolerant condition. However, factors contributing to the increased VMH NE activity remain unknown. This study therefore investigated in normal rats the effect of a hyperinsulinemic-euglycemic clamp on VMH monoamine turnover and utilization via simultaneous VMH microdialysis to establish a role for hyperinsulinemia in the stimulation of VMH NE activity. Within 20 min of initiation of the hyperinsulinemic-euglycemic clamp, VMH extracellular methoxyhydroxy phenylglycol (metabolite of NE) level increased by 54% and remained approximately at this level for the 100 min duration of the clamp relative to control values (p<0.05). Hyperinsulinemia did not affect VMH dopamine or serotonin metabolism. Subsequent establishment of a hyperinsulinemic-hypoglycemic camp did not alter the VMH monoamine metabolism profile relative to the hyperinsulinemic-euglycemic clamp. Infusion of saline (as control) in a separate group of rats over the entire clamp period induced no changes in any monoamine metabolic profile relative to baseline. Hyperinsulinemia can feedback to stimulate VMH NE activity and, as a result, may contribute to the initiation and/or perpetuation of the obese, hyperinsulinemic, glucose-intolerant state.


Subject(s)
Hyperinsulinism/blood , Norepinephrine/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Blood Glucose/metabolism , Glucose Clamp Technique , Homovanillic Acid/metabolism , Hydroxyindoleacetic Acid/metabolism , Insulin/blood , Male , Methoxyhydroxyphenylglycol/metabolism , Microdialysis , Rats , Rats, Sprague-Dawley , Wakefulness/physiology
9.
Am J Physiol Regul Integr Comp Physiol ; 278(2): R435-44, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666145

ABSTRACT

Increases in ventromedial hypothalamic (VMH) norepinephrine (NE) levels and/or activities have been observed in a variety of animal models of the obese insulin-resistant condition. This study examined the metabolic effects of chronic NE infusion (25 nmol/h) into the unilateral VMH of normal rats. Within 4 days, VMH NE infusion significantly increased plasma insulin (140%), glucagon (45%), leptin (300%), triglyceride (100%), abdominal fat pad weight (50%), and white adipocyte lipogenic (100%) and lipolytic (100%) activities relative to vehicle-infused rats. Furthermore, isolated islet insulin secretory response to glucose (15 mM) within 4 days of such treatment was increased over twofold (P < 0.05). Among treated animals, fat stores continued to increase over time and plateaued at approximately 2 wk (3-fold increase), remaining elevated to the end of the study (5 wk). By week 4 of treatment, NE infusion induced glucose intolerance as evidenced by a 32% increase in plasma glucose total area under the glucose tolerance test curve (P < 0.01). Whole body fat oxidation rate measured after 5 wk of infusion was significantly increased among treated animals as evidenced by a reduced respiratory quotient (0.87 +/- 0.01) relative to controls (0. 90 +/- 0.01). VMH NE infusion induced hyperphagia (30%) only during the first week and did not affect body weight over the 5-wk period. Increases in VMH NE activity that are common among obese insulin-resistant animal models can cause the development of this obese glucose-intolerant (metabolic) syndrome.


Subject(s)
Glucose Intolerance , Norepinephrine/administration & dosage , Obesity/physiopathology , Ventromedial Hypothalamic Nucleus/physiology , Adipose Tissue/pathology , Animals , Carbohydrate Metabolism , Endocrine Glands/physiopathology , Female , Hormones/blood , Injections , Isoproterenol/pharmacology , Leptin/blood , Lipid Metabolism , Lipolysis/drug effects , Lipolysis/physiology , Norepinephrine/pharmacology , Obesity/pathology , Rats , Rats, Sprague-Dawley , Reference Values , Syndrome , Time Factors
10.
Neuroendocrinology ; 71(1): 68-78, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10644901

ABSTRACT

Hypothalamic neuropeptide Y (NPY) and corticotropin-releasing hormone (CRH) influence feeding and levels of plasma glucose, insulin, free fatty acids, and triglycerides. Treatment of genetically obese, ob/ob mice, with dopamine receptor D(1)/D(2) agonists normalizes hyperphagia, body weight gain, hyperglycemia, and hyperlipidemia. We therefore examined whether levels of NPY and CRH immunoreactivity in discrete hypothalamic nuclei are altered in ob/ob mice, and whether dopaminergic treatment reverses this alteration. Female ob/ob mice were treated daily at 1 h after light onset with the D(1)/D(2) agonists, SKF-38393 (20 mg/kg) and bromocriptine (15 mg/kg), respectively or vehicle for 2 weeks. Such treatment, while normalizing body weight gain and hyperglycemia, also significantly reduced elevated NPY immunoreactivity in the suprachiasmatic (by 39%), intergeniculate (by 43%), paraventricular (PVN; by 31%), and arcuate (by 41%) nuclei in obese mice to levels observed in lean mice. This treatment also caused a 45-50% decline in levels of CRH in the PVN and dorsomedial hypothalamus compared to obese controls to levels observed in lean mice. Taken together, these findings suggest that dopaminergic D(1)/D(2) receptor coactivation may improve hyperphagia, hyperglycemia, and obesity in the ob/ob mouse, in part, by normalizing elevated levels of both NPY and CRH.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Dopamine D2 Receptor Antagonists , Hyperglycemia/metabolism , Hypothalamus/metabolism , Neuropeptide Y/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Blood Glucose , Corticotropin-Releasing Hormone/genetics , Dorsomedial Hypothalamic Nucleus/chemistry , Dorsomedial Hypothalamic Nucleus/drug effects , Dorsomedial Hypothalamic Nucleus/metabolism , Eating , Female , Gene Expression/drug effects , Hypothalamus/chemistry , Hypothalamus/drug effects , Mice , Mice, Inbred C57BL , Mice, Obese , Neuropeptide Y/genetics , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/analysis , Suprachiasmatic Nucleus/chemistry , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Weight Gain
11.
Metabolism ; 48(10): 1287-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10535392

ABSTRACT

To examine the possibility of a cause-effect relationship between enhanced monoamine content in the ventromedial hypothalamus ([VMH] a characteristic of hyperinsulinemic and insulin-resistant animals) and islet dysfunction, we infused norepinephrine ([NE] 25 nmol/h) and/or serotonin ([5-HT] 2.5 nmol/h) into the VMH of normal hamsters for 5 weeks and then examined insulin release from the isolated pancreatic islets. VMH infusion of NE + 5-HT, but not of either neurotransmitter alone, produced a marked leftward shift in the dose-response curve of glucose-induced insulin release (twofold to sixfold increase at 5 to 7.5 mmol/L glucose v vehicle-treated animals). In addition, the islet responsiveness to 1 micromol/L NE and 10 micromol/L acetylcholine was abolished in these NE + 5-HT VMH-infused hamsters. These findings indicate that an increase of NE and 5-HT content in the VMH can induce dysregulation of islet insulin release in response to glucose and neurotransmitters. Inasmuch as VMH NE and 5-HT levels are elevated in hyperinsulinemic and insulin-resistant animals, the present findings suggest that an endogenous increase in these hypothalamic monoamines may contribute to islet dysfunction, which is one of the characteristics of type 2 diabetes.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Norepinephrine/pharmacology , Serotonin/pharmacology , Ventromedial Hypothalamic Nucleus/physiology , Acetylcholine/pharmacology , Animals , Cricetinae , Dose-Response Relationship, Drug , Drug Interactions , Glucose/pharmacology , In Vitro Techniques , Infusions, Parenteral , Insulin Secretion , Islets of Langerhans/drug effects , Male , Mesocricetus , Norepinephrine/administration & dosage , Serotonin/administration & dosage , Ventromedial Hypothalamic Nucleus/drug effects
12.
Metabolism ; 48(8): 1033-40, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10459570

ABSTRACT

Our previous studies have shown that the dopaminergic D1 receptor agonist SKF38393 (SKF) plus the D2 receptor agonist bromocriptine (BC) act synergistically to reduce obesity in obese C57BL/6J (ob/ob) mice. The present study investigated the effects of this combination on dyslipidemia in ob/ob mice. Female ob/ob mice were treated daily with vehicle or SKF (20 mg/kg body weight [BW]) plus BC (16 mg/kg BW [BC/SKF]) for 14 days. The animals were then used for the characterization of plasma lipoprotein profiles, hepatic triacylglycerol synthesis and secretion, adipocyte lipolysis, adipose and muscle lipoprotein lipase (LPL) activity, and muscle triglyceride (TG) content. The treatment significantly reduced plasma glucose 54%, TG 41%, cholesterol 21%, phospholipid 20%, and free fatty acid (FFA) 36% (P < .01). Hepatic triacylglycerol synthesis was 55% lower in treated mice versus control mice (P < .01). The cell size of isolated adipocytes was significantly reduced (41%) by treatment. LPL activity was increased in soleus skeletal muscle (25%, P < .05) but was sharply reduced in adipose tissue (91%, P < .01) in treated versus control mice. The TG content of hindlimb muscle was about 49% lower in treated versus control mice (P < .05). The basal and isoproterenol-stimulated lipolytic rate was decreased (approximately 53%) in adipocytes from treated animals compared with the control (P < .01). In conclusion, BC/SKF normalized the hypertriglyceridemia likely via its simultaneous antilipogenic action in liver tissue and antilipolytic action in adipose tissue. Decreased plasma flux of FFA partially contributed to the reduced hepatic lipogenesis, plasma very-low-density lipoprotein (VLDL)-TG, and TG in skeletal muscle. The above-described effects of BC/SKF treatment are largely independent of its effect to normalize hyperphagia in ob/ob mice.


Subject(s)
2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/therapeutic use , Bromocriptine/therapeutic use , Dopamine Agonists/therapeutic use , Hyperlipidemias/drug therapy , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Adipose Tissue/metabolism , Animals , Bromocriptine/pharmacology , Dopamine Agonists/pharmacology , Drug Synergism , Drug Therapy, Combination , Female , Hyperlipidemias/metabolism , Lipolysis/drug effects , Lipoproteins/blood , Mice , Mice, Obese , Receptors, Dopamine/metabolism , Triglycerides/metabolism
13.
Neuroreport ; 10(10): 2073-7, 1999 Jul 13.
Article in English | MEDLINE | ID: mdl-10424677

ABSTRACT

A critical role for temporal organization of dopaminergic and serotonergic activities within the suprachiasmatic nuclei (SCN) in the regulation of peripheral glucose metabolism has been postulated. This study employed in vivo microdialysis to investigate the temporal extracellular profiles of dopamine and serotonin metabolites in the SCN of freely behaving naturally glucose tolerant and intolerant Syrian hamsters. Microdialysis samples from the right SCN of awake, glucose tolerant or intolerant hamsters held on 14 h daily photoperiods were collected every 2 h over a 24 h period and assayed via HPLC for the metabolites of dopamine: homovanillic acid (HVA) and serotonin (5-hydroxyindolacetic acid, 5-HIAA). Among glucose tolerant hamsters, daily rhythms of SCN HVA and 5-HIAA were observed with coincident plateaus throughout the nocturnal phase of the day (both p<0.01). Relative to glucose tolerant hamsters, glucose intolerant animals exhibited a loss in the daily rhythm of SCN HVA (p<0.0001) and 5-HIAA (p<0.02) due to marked reductions (70%) throughout the 24 h period in HVA levels and comparative decreases (35%) in nocturnal peak levels of 5-HIAA. These findings demonstrate that daily profiles of extracellular dopamine and serotonin activities in the SCN, known to influence glucose metabolism, differ between glucose tolerant and intolerant hamsters.


Subject(s)
Biogenic Monoamines/metabolism , Glucose Intolerance/metabolism , Suprachiasmatic Nucleus/metabolism , Analysis of Variance , Animals , Circadian Rhythm/physiology , Cricetinae , Dopamine/metabolism , Glucose Tolerance Test , Male , Mesocricetus , Microdialysis , Serotonin/metabolism
14.
Chronobiol Int ; 16(3): 315-33, 1999 May.
Article in English | MEDLINE | ID: mdl-10373101

ABSTRACT

In the present study, we investigated the time-dependent interactive effects of daily injections of prolactin (PRL) and corticosterone (CORT) on the activation of lymphocyte function and inhibition of tumor growth in vivo in mice. BALB/c mice were injected subcutaneously with EMT-6 fibrosarcoma cells (a murine connective tissue tumor cell derived from mammary gland), and then different groups of animals were treated with PRL (1 microg/g body weight [BW] ip) at Oh, 4h, 8h, 12h, 16h, or 20h after CRT (1 microg/g BW ip) daily for 10 days. Different control groups were vehicle treated or treated with either hormone alone. Mice were kept in constant light 1 week before and during injections and in a 14:10 light-dark cycle thereafter. Tumor progression was monitored for up to 21 days after the cessation of treatment, and thereafter spleen lymphocytes were harvested and tested for mitogen-triggered proliferation. Prolactin administration at 8h or 16-20h after corticosteroid treatment reduced tumor volume by 77% and 49%, respectively, relative to vehicle-treated controls. Other time relations of hormone treatment were ineffectual. Further studies indicated that the immunosuppressant cyclosporin A (CSA) substantially stimulated tumor growth; this effect was completely abrogated by a simultaneous 8h related hormone treatment. How ever, the 8h hormone treatment was ineffective in inhibiting tumor growth in T-cell-deficient nude mice. Spleen lymphocytes from tumor-bearing (TB) mice showed an elevated basal proliferative capacity stimulated by concanavalin A (ConA; a stimulus for T-cell proliferation) and lipopolysaccharide (LPS; a stimulus for B-cell proliferation) compared to non-TB mice. Spleen lymphocytes from TB mice treated with CORT and PRL at 8h intervals exhibited an increased spontaneous (as well as LPS- and ConA- triggered) proliferation (by 104%, 48%, and 70%, respectively) compared with vehicle control TB mice. Fluorescence-activated cell sorting (FACS) analysis of splenocytes from hormone-treated animals indicated a 34-100% increase in the CD4+ (e.g., T helper cell) population. Treatment of animals with either hormone alone did not inhibit tumor growth or stimulate immune function relative to vehicle controls. The daily rhythms of plasma PRL, CORT, and thyroxine were all substantially altered by the presence of tumor in these mice. These results indicate that appropriately timed daily treatment of PRL and CORT can attenuate tumor growth, in part, via activation of antitumor immune mechanisms. Collectively, these data suggest that circadian neuroendocrine activities must be temporally organized appropriately to inhibit tumor growth.


Subject(s)
Corticosterone/pharmacology , Fibrosarcoma/pathology , Lymphocytes/immunology , Photoperiod , Prolactin/pharmacology , Sarcoma, Experimental/pathology , Animals , Biological Clocks , Cell Division/drug effects , Circadian Rhythm , Corticosterone/administration & dosage , Darkness , Drug Administration Schedule , Fibrosarcoma/drug therapy , Fibrosarcoma/immunology , Light , Lymphocytes/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prolactin/administration & dosage , Prolactin/blood , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/immunology , Spleen/immunology , Time Factors
15.
Photochem Photobiol ; 69(5): 575-81, 1999 May.
Article in English | MEDLINE | ID: mdl-10333764

ABSTRACT

The role of the host immune system in contributing to tumor regression following benzophenothiazine photodynamic therapy (PDT) was examined. Photodynamic therapy with 2-iodo-5-ethylamino-9-diethylaminobenzo[a]-phenothiazinium chloride (2I-EtNBS) eradicated EMT-6 mammary fibrosarcomas in 75-100% of treated mice. In contrast, PDT failed to inhibit tumor growth in T-cell-deficient nude mice. Furthermore, T-cell depletion studies with anti-CD8 antibody revealed that the CD8+ T-cell population was critical for an effective PDT response (tumor volume 14 days post-PDT: 262 mm3 vs 59 mm3 in controls; P < 0.01). Because anti-CD4 antibody inhibited tumor growth in the absence of PDT, the role of CD4+ T cells remains unclear. Depletion of natural killer (NK) cells in vivo with anti-asialo-GM1 antibody significantly reduced a suboptimal PDT effect relative to vehicle controls (tumor volume 13 days post-PDT: 513 mm3 vs 85 mm3, respectively; P < 0.001). However, splenic NK cells obtained from PDT-treated tumor-bearing mice were not cytotoxic in vitro against EMT-6 cells, suggesting that NK cells contribute to the PDT effect in vivo by an indirect mechanism. In addition, when mice with complete tumor regression following PDT were rechallenged 28 days later with 5 x 10(5) EMT-6 cells, tumor growth was significantly inhibited as compared to controls (tumor volume 40 days postrechallenge: 137 mm3 vs 833 mm3 in controls; P < 0.03; percent animals without tumor in five experiments: 67% vs 8% in controls). Collectively, these results demonstrate that CD8+ T cells are required to prevent tumor regrowth after 2I-EtNBS-PDT, NK cells contribute to this response and such PDT can elicit protective antitumor immunity.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms, Experimental/drug therapy , Photochemotherapy , Thiazines/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , CD8-Positive T-Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/immunology , Thiazines/pharmacokinetics
16.
Int J Obes Relat Metab Disord ; 23(4): 425-31, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10340822

ABSTRACT

OBJECTIVE: We previously reported that a two week treatment with SKF 38393 (SKF, a dopamine D1 receptor agonist), plus bromocriptine (BC, a dopamine D2 receptor agonist) acted synergistically to normalize hyperphagia, body fat, hyperglycaemia and hyperlipidaemia in ob/ob mice. The present study further investigates the biochemical mechanisms triggered by this drug treatment. DESIGN: Six week old female C57BL/6J ob/ob mice were divided into three groups and treated for two weeks with either BC and SKF, vehicle (control), or vehicle and pair fed to match the drug-treated group's daily food intake. RESULTS: BC/SKF treatment reduced food consumption by 55%, and treated mice weighed less than either pair fed or ad libitum fed controls after two weeks of treatment. Moreover, oxygen consumption was increased by 2.4-fold and the respiratory quotient (RQ) decreased from 1.23 to 0.96 (indicating a reduction in de novo lipogenesis) by drug treatment relative to ad libitum fed controls, but these parameters were unaffected by pair feeding control mice. The treatment also reduced blood glucose and free fatty acids (FFA) relative to pair fed and ad libitum fed controls. BC/SKF treatment (but not pair feeding) concurrently reduced lipolysis, lipogenic enzyme activities and hepatic gluconeogenic enzyme activities. Treatment also increased hepatic concentrations of glycogen and xylulose-5-phosphate (X-5-P), a key stimulator of glycolysis. Finally, BC/SKF, but not pair feeding, reduced the circulating concentrations of thyroxine and corticosterone, two hormones known to increase lipolysis, lipogenesis and hyperglycaemia. Drug treatment also increased serum dehydroepiandrosterone (DHEA) sulfate concentrations, an inhibitor of body fat store accumulation. CONCLUSION: These findings demonstrate that BC/SKF treatment not only normalizes hyperphagia of ob/ob mice, but also redirects several metabolic and endocrine activities, independent of its effects on feeding to improve the obese-diabetic syndrome in ob/ob mice.


Subject(s)
Diabetes Mellitus/drug therapy , Dopamine Agonists/therapeutic use , Obesity , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/administration & dosage , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Bromocriptine/administration & dosage , Bromocriptine/therapeutic use , Drug Synergism , Eating/drug effects , Energy Metabolism/drug effects , Fatty Acids, Nonesterified/blood , Female , Gluconeogenesis/drug effects , Lipids/biosynthesis , Lipolysis/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Weight Loss
17.
Neuroendocrinology ; 69(3): 160-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10087448

ABSTRACT

Bromocriptine, a potent dopamine D2 receptor agonist, suppresses lipogenesis and improves glucose intolerance and insulin resistance. Recent evidence suggests that bromocriptine may produce these effects by altering central nervous system (CNS) regulation of metabolism. To determine whether or not the CNS plays a critical role in these bromocriptine-mediated effects on peripheral metabolism, we compared the metabolic responses to bromocriptine when administered peripherally versus centrally in naturally obese and glucose intolerant Syrian hamsters. Male hamsters (BW 194 +/- 5 g) were treated with bromocriptine or vehicle either intraperitoneally (i.p., 800 microgram/animal) or intracerebroventricularly (i.c.v., 1 microgram/animal) daily at 1 h after light onset for 14 days while held on 14-hour daily photoperiods. Glucose tolerance tests (GTTs, 3 g glucose/kg BW) were conducted after treatment. Compared to control animals, bromocriptine i.p. significantly reduced weight gain (11.7 vs. -2.4 g) and the areas under the glucose and insulin GTT curves by 29 and 48%, respectively. Similarly, compared with vehicle-treated controls, bromocriptine i.c.v. at 1 microgram/animal substantially reduced weight gain (8.7 vs. -6.3 g), the areas under the glucose and insulin GTT curves by 31 and 44% respectively, and the basal plasma insulin concentration by 41% (p < 0.05). Furthermore, both treatments significantly improved insulin-mediated suppression of hepatic glucose production during a hyperinsulinemic-euglycemic clamp. Thus, daily administration of bromocriptine at a very low dose i.c.v. replicates the metabolic effects of bromocriptine administered i.p. at a much higher dose. This finding demonstrates for the first time that the CNS is a critical target of bromocriptine's metabolic effects.


Subject(s)
Bromocriptine/administration & dosage , Glucose Intolerance , Insulin Resistance , Animals , Blood Glucose/metabolism , Bromocriptine/pharmacology , Bromocriptine/therapeutic use , Cricetinae , Glucose Clamp Technique , Injections, Intraventricular , Insulin/blood , Male , Mesocricetus , Obesity/drug therapy , Obesity/physiopathology , Photoperiod , Weight Gain/drug effects
18.
Neuroendocrinology ; 70(6): 460-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10657739

ABSTRACT

The ventromedial hypothalamus (VMH) is involved in the regulation of peripheral metabolism. We and others have shown that activities, or extracellular metabolites of norepinephrine (NE) and serotonin (5-HT) are elevated in the VMH of both genetically and seasonally insulin-resistant and glucose-intolerant animals. This study examined whether chronic increases in VMH NE and 5-HT concentration of normal animals might lead to insulin-resistant and glucose-intolerant conditions in hamsters. Euinsulinemic, glucose-tolerant hamsters were infused continuously for 5 weeks into the right VMH with either vehicle, NE (5 or 25 nmol/h), 5-HT (2.5 nmol/h), or NE (5 or 25 nmol/h) plus 5-HT (2.5 nmol/h) through osmotic minipumps. Compared to vehicle, NE (25 nmol/h) significantly increased the glucose total area under the curve (TAUC) by 32% during glucose tolerance tests (GTT) conducted after 5 weeks' infusion. 5-HT alone significantly increased the GTT insulin TAUC (131%) and basal plasma insulin level (116%) but not glucose TAUC. NE (5 nmol/h) plus 5-HT infusion significantly increased insulin TAUC (129%) and basal plasma insulin (120%), whereas NE (25 nmol/h) plus 5-HT infusion significantly increased both the GTT glucose and insulin TAUC (43 and 113%, respectively), as well as basal plasma insulin level (158%), relative to vehicle infusion. Our findings demonstrate for the first time the differential and, more importantly, interactive effects of increased VMH NE and 5-HT in producing hyperinsulinemia, insulin resistance and glucose intolerance.


Subject(s)
Blood Glucose/metabolism , Insulin Resistance , Norepinephrine/pharmacology , Serotonin/pharmacology , Sympathomimetics/pharmacology , Ventromedial Hypothalamic Nucleus/drug effects , Animals , Body Weight , Cricetinae , Glucose Tolerance Test , Hyperinsulinism/chemically induced , Insulin/blood , Male , Mesocricetus
19.
Expert Opin Investig Drugs ; 8(10): 1683-1707, 1999 10.
Article in English | MEDLINE | ID: mdl-11139820

ABSTRACT

Bromocriptine, a potent dopamine D(2) receptor agonist, has been shown to reduce insulin resistance, glucose intolerance and hyperlipidaemia in both numerous animal studies and in Phase II studies. Bromocriptine has been used worldwide for over 20 years to treat Parkinson's disease, macroprolactinoma and other disorders; it has been found to be generally safe. We therefore investigated the possible beneficial effects of Ergoset(R) (Ergo Science Corp.), a new quick release formulation of bromocriptine, on glycaemic control and serum lipid profile in obese Type 2 diabetic subjects in two large Phase III studies. A large, randomised, double-blind placebo-controlled study was conducted in which Ergoset was given once daily at 8 am. (4.8 mg maximum dose) for 24 weeks as adjunctive therapy to sulphonylurea (485 subjects) to obese Type 2 diabetics held on a weight- maintaining diet. Treatment efficacy parameters included change from baseline in glycated haemoglobin A(1c) (HbA(1c)), fasting and post-prandial serum glucose, insulin, triglyceride and free fatty acid levels. Baseline glycated haemoglobin, fasting glucose, insulin, triglyceride and free fatty acid levels did not differ between treatment groups. and on average were 9.4 +/- 0.05%, 222 +/- 2 mg/dl, 24 +/- 1 µU/ml, 248 +/- 11 mg/dl, and 850 +/- 32 µEq/l, respectively. A similarly designed study of Ergoset as monotherapy in Type 2 diabetics (154 subjects) with similar baseline clinical characteristics was conducted. Addition of Ergoset treatment to sulphonylurea reduced percent glycated HbA(1c) by 0.55 (P < 0.0001) (approximately 1.0 for responders, 65% of population), fasting and post-prandial glucose by 23 and 26 mg/dl (P < 0.0002), fasting and post-prandial triglycerides by 72 and 63 mg/dl (P < 0.005) and fasting and post-prandial free fatty acids by 150 and 165 µEq/l (P < 0.05), relative to placebo. Twelve percent of all Ergoset subjects, compared to 3% of placebo subjects, withdrew from the study due to adverse events. The most common events causing withdrawal were nausea, dizziness, asthenia, and rhinitis (representing 4.5, 3.3, 2.0, and 0.8% of the total Ergoset populations, respectively). The incidence of serious adverse events did not differ between Ergoset- (3.4%) and placebo- (4.3%) treated subjects. Ergoset as monotherapy also improved glycaemic control (0.56 HbA(1c) decrease relative to placebo after 24 weeks of treatment; P < 0.02). Once daily Ergoset treatment improves glycaemic control and serum lipid profile and is well-tolerated in obese Type 2 diabetics.

20.
Biochim Biophys Acta ; 1405(1-3): 1-13, 1998 Oct 21.
Article in English | MEDLINE | ID: mdl-9784590

ABSTRACT

One of the characteristics of obesity-associated diabetes is an elevated fasting plasma insulin concentration with a weak insulin secretory response to subsequent glucose stimulation. Evidence suggests that hyperglycemia and hyperlipidemia may contribute to the initiation and progression of this disordered islet glucose sensing. It has been proposed that reducing hyperglycemia and hyperlipidemia per se may improve islet glucose sensing. Here we studied glucose-dependent insulin release in islets isolated from ob/ob mice treated with dopamine agonists (bromocriptine and SKF38393, BC/SKF) which significantly reduced circulating glucose and lipid levels of ob/ob mice. Islets from BC/SKF-treated mice showed a marked decrease of the elevated basal insulin release to levels similar to lean mice. Such treatment also induced a higher secretory response to glucose stimulation compared with that in ob/ob mice with sustained hyperglycemia and hyperlipidemia. Similarly, when islets from untreated ob/ob mice were cultured for 7 days in 11 mM glucose in the absence of free fatty acid, the basal insulin release was significantly decreased and high glucose stimulated insulin release increased compared with that from islets cultured in medium containing 30 mM glucose and 2 mM oleate. The BC/SKF-induced reduction of elevated basal insulin release was associated with decreased hexokinase activity and basal cyclic AMP content in islet tissue. Our results demonstrate that dopamine agonist treatment improves basal insulin release in ob/ob mice and this effect may be mediated, in part, by a reduction of hyperglycemia and hyperlipidemia.


Subject(s)
Dopamine Agonists/pharmacology , Hyperglycemia/prevention & control , Hyperlipidemias/prevention & control , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Blood Glucose/metabolism , Bromocriptine/pharmacology , Colforsin/pharmacology , Cyclic AMP/metabolism , Glucokinase/metabolism , Glucose/pharmacology , Hexokinase/metabolism , Hyperglycemia/blood , Hyperglycemia/genetics , Hyperlipidemias/blood , Hyperlipidemias/genetics , In Vitro Techniques , Insulin Secretion , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Obese , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL