Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Rep ; 30(6): 2028-2039.e4, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049029

ABSTRACT

The vagus nerve conveys gastrointestinal cues to the brain to control eating behavior. In obesity, vagally mediated gut-brain signaling is disrupted. Here, we show that the cocaine- and amphetamine-regulated transcript (CART) is a neuropeptide synthesized proportional to the food consumed in vagal afferent neurons (VANs) of chow-fed rats. CART injection into the nucleus tractus solitarii (NTS), the site of vagal afferent central termination, reduces food intake. Conversely, blocking endogenous CART action in the NTS increases food intake in chow-fed rats, and this requires intact VANs. Viral-mediated Cartpt knockdown in VANs increases weight gain and daily food intake via larger meals and faster ingestion rate. In obese rats fed a high-fat, high-sugar diet, meal-induced CART synthesis in VANs is blunted and CART antibody fails to increase food intake. However, CART injection into the NTS retains its anorexigenic effect in obese rats. Restoring disrupted VAN CART signaling in obesity could be a promising therapeutic approach.


Subject(s)
Hyperphagia/genetics , Nerve Tissue Proteins/metabolism , Vagus Nerve/drug effects , Weight Gain/genetics , Animals , Humans , Male , Rats
3.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R708-R720, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29847161

ABSTRACT

Endogenous intestinal glucagon-like peptide-1 (GLP-1) controls satiation and glucose metabolism via vagal afferent neurons (VANs). Recently, VANs have received increasing attention for their role in brown adipose tissue (BAT) thermogenesis. It is, however, unclear whether VAN GLP-1 receptor (GLP-1R) signaling affects BAT thermogenesis and energy expenditure (EE) and whether this VAN mechanism contributes to energy balance. First, we tested the effect of the GLP-1R agonist exendin-4 (Ex4, 0.3 µg/kg ip) on EE and BAT thermogenesis and whether these effects require VAN GLP-1R signaling using a rat model with a selective Glp1r knockdown (kd) in VANs. Second, we examined the role of VAN GLP-1R in energy balance during chronic high-fat diet (HFD) feeding in VAN Glp1r kd rats. Finally, we used viral transsynaptic tracers to identify the possible neuronal substrates of such a gut-BAT interaction. VAN Glp1r kd attenuated the acute suppressive effects of Ex4 on EE and BAT thermogenesis. Consistent with this finding, the VAN Glp1r kd increased EE and BAT activity, diminished body weight gain, and improved insulin sensitivity compared with HFD-fed controls. Anterograde transsynaptic viral tracing of VANs infected major hypothalamic and hindbrain areas involved in BAT sympathetic regulation. Moreover, retrograde tracing from BAT combined with laser capture microdissection revealed that a population of VANs expressing Glp1r is synaptically connected to the BAT. Our findings reveal a novel role of VAN GLP-1R signaling in the regulation of EE and BAT thermogenesis and imply that through this gut-brain-BAT connection, intestinal GLP-1 plays a role in HFD-induced metabolic syndrome.


Subject(s)
Adipose Tissue, Brown/innervation , Autonomic Nervous System/metabolism , Brain/metabolism , Energy Metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Intestines/innervation , Thermogenesis , Animals , Autonomic Nervous System/drug effects , Diet, High-Fat , Energy Metabolism/drug effects , Exenatide/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Incretins/pharmacology , Male , Neural Pathways/metabolism , Neurons, Afferent/metabolism , Rats, Sprague-Dawley , Signal Transduction , Thermogenesis/drug effects
4.
Physiol Behav ; 192: 194-199, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29654813

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) secreted from intestinal L-cells plays a major role in meal termination and glucose-dependent insulin secretion. Several lines of evidence indicate, however, that the acute satiating and incretin effects of GLP-1 are attenuated with high fat diet (HFD) exposure. Here we tested the hypothesis that endogenous GLP-1 differentially affects energy balance and glucose homeostasis dependent on whether rats are fed chow or HFD (60% energy from fat). METHODS: We blocked GLP-1 receptor (GLP-1R) signaling by daily intraperitoneal (IP) injection of the GLP-1R antagonist exendin (9-39) (Ex9, 10 µg/kg) or vehicle for 5 weeks in male Sprague-Dawley rats fed either chow or HFD, recorded body weight (BW) and food intake throughout, and assessed energy expenditure (3rd week) and glucose tolerance (4th week). RESULTS: Five week daily Ex9 injections reduced BW gain in HFD-fed rats, but did not affect BW in chow-fed rats. On the other hand, chronic Ex9 treatment did not affect daily food intake in either chow or HFD-fed rats during the entire study. The reduced BW gain in HFD-fed rats was associated with an increase in energy expenditure. Interestingly, chronic Ex9 treatment induced glucose intolerance in chow-fed rats, but not in HFD-fed rats, suggesting a differential role of GLP-1R signaling in glucose metabolism during chow and HFD feeding. CONCLUSIONS: Our findings reveal a novel role of GLP-1R signaling, modulating energy expenditure rather than eating behavior during HFD feeding. Furthermore, these results suggest a previously unrecognized contribution of GLP-1R signaling to the pathophysiology of obesity.


Subject(s)
Diet, High-Fat/adverse effects , Energy Metabolism/physiology , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Anti-Obesity Agents/pharmacology , Body Composition/drug effects , Body Composition/physiology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucose/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Male , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Weight Gain/drug effects , Weight Gain/physiology
5.
Mol Metab ; 11: 33-46, 2018 05.
Article in English | MEDLINE | ID: mdl-29650350

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) neurons in the hindbrain densely innervate the dorsomedial hypothalamus (DMH), a nucleus strongly implicated in body weight regulation and the sympathetic control of brown adipose tissue (BAT) thermogenesis. Therefore, DMH GLP-1 receptors (GLP-1R) are well placed to regulate energy balance by controlling sympathetic outflow and BAT function. METHODS: We investigate this possibility in adult male rats by using direct administration of GLP-1 (0.5 ug) into the DMH, knocking down DMH GLP-1R mRNA with viral-mediated RNA interference, and by examining the neurochemical phenotype of GLP-1R expressing cells in the DMH using in situ hybridization. RESULTS: GLP-1 administered into the DMH increased BAT thermogenesis and hepatic triglyceride (TG) mobilization. On the other hand, Glp1r knockdown (KD) in the DMH increased body weight gain and adiposity, with a concomitant reduction in energy expenditure (EE), BAT temperature, and uncoupling protein 1 (UCP1) expression. Moreover, DMH Glp1r KD induced hepatic steatosis, increased plasma TG, and elevated liver specific de-novo lipogenesis, effects that collectively contributed to insulin resistance. Interestingly, DMH Glp1r KD increased neuropeptide Y (NPY) mRNA expression in the DMH. GLP-1R mRNA in the DMH, however, was found in GABAergic not NPY neurons, consistent with a GLP-1R-dependent inhibition of NPY neurons that is mediated by local GABAergic neurons. Finally, DMH Glp1r KD attenuated the anorexigenic effects of the GLP-1R agonist exendin-4, highlighting an important role of DMH GLP-1R signaling in GLP-1-based therapies. CONCLUSIONS: Collectively, our data show that DMH GLP-1R signaling plays a key role for BAT thermogenesis and adiposity.


Subject(s)
Adipose Tissue, Brown/metabolism , Adiposity , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus/metabolism , Thermogenesis , Animals , Exenatide/metabolism , GABAergic Neurons/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Insulin Resistance , Lipogenesis , Male , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Uncoupling Protein 1/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R811-R823, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29384699

ABSTRACT

Hindbrain catecholamine neurons convey gut-derived metabolic signals to an interconnected neuronal network in the hypothalamus and adjacent forebrain. These neurons are critical for short-term glycemic control, glucocorticoid and glucoprivic feeding responses, and glucagon-like peptide 1 (GLP-1) signaling. Here we investigate whether these pathways also contribute to long-term energy homeostasis by controlling obesogenic sensitivity to a high-fat/high-sucrose choice (HFSC) diet. We ablated hindbrain-originating catecholaminergic projections by injecting anti-dopamine-ß-hydroxylase-conjugated saporin (DSAP) into the paraventricular nucleus of the hypothalamus (PVH) of male rats fed a chow diet for up to 12 wk or a HFSC diet for 8 wk. We measured the effects of DSAP lesions on food choices; visceral adiposity; plasma glucose, insulin, and leptin; and indicators of long-term ACTH and corticosterone secretion. We also determined lesion effects on the number of carbohydrate or fat calories required to increase visceral fat. Finally, we examined corticotropin-releasing hormone levels in the PVH and arcuate nucleus expression of neuropeptide Y ( Npy), agouti-related peptide ( Agrp), and proopiomelanocortin ( Pomc). DSAP-injected chow-fed rats slowly increase visceral adiposity but quickly develop mild insulin resistance and elevated blood glucose. DSAP-injected HFSC-fed rats, however, dramatically increase food intake, body weight, and visceral adiposity beyond the level in control HFSC-fed rats. These changes are concomitant with 1) a reduction in the number of carbohydrate calories required to generate visceral fat, 2) abnormal Npy, Agrp, and Pomc expression, and 3) aberrant control of insulin secretion and glucocorticoid negative feedback. Long-term metabolic adaptations to high-carbohydrate diets, therefore, require intact forebrain catecholamine projections. Without them, animals cannot alter forebrain mechanisms to restrain increased visceral adiposity.


Subject(s)
Catecholamines/metabolism , Nerve Net/physiopathology , Obesity/physiopathology , Prosencephalon/physiopathology , Animals , Blood Glucose/metabolism , Diet , Dopamine beta-Hydroxylase/antagonists & inhibitors , Energy Intake , Insulin/blood , Intra-Abdominal Fat/drug effects , Leptin/blood , Male , Neural Pathways/physiopathology , Paraventricular Hypothalamic Nucleus , Rats , Rats, Sprague-Dawley , Saporins/pharmacology
7.
Mol Metab ; 5(7): 552-565, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27408779

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) analogs are attractive options for the treatment of type II diabetes and obesity because of their incretin and anorexigenic effects. Peripheral administration of the GLP-1R agonist Exendin-4 (Ex-4) also increases glucocorticoid secretion in rodents and humans, but whether the released glucocorticoids interact with Ex-4's anorexigenic effect remains unclear. METHODS: To test this, we used two experimental approaches that suppress corticosterone secretion and then assessed Ex-4 effects on eating in adult male rats. First, we combined acute and chronic low dose dexamethasone treatment with Ex-4. Second, we ablated hindbrain catecholamine neurons projecting to the hypothalamus with anti-dopamine-ß-hydroxylase-saporin (DSAP) to block Ex-4-induced corticosterone secretion. RESULTS: Combining dexamethasone and Ex-4 produced a larger acute anorexigenic effect than Ex-4 alone. Likewise, chronic dexamethasone and Ex-4 co-treatment produced a synergistic effect on eating and greater body weight loss in diet-induced obese rats than Ex-4 alone. DSAP lesions not only blunted Ex-4's ability to increase corticosterone secretion, but potentiated the anorexigenic effect of Ex-4, indicating that Ex-4-dependent corticosterone secretion opposes Ex-4's actions. Consistent with the enhancement of Ex-4's anorexigenic effect, DSAP lesion altered Ex-4-dependent changes in neuropeptide Y, preproglucagon, and corticotropin releasing hormone gene expression involved in glucocorticoid feedback. CONCLUSIONS: Our findings demonstrate that limiting glucocorticoid secretion and actions with low dose dexamethasone or DSAP lesion increases Ex-4's ability to reduce food intake and body weight. Novel glucocorticoid receptor based mechanisms, therefore, may help enhance GLP-1-based obesity therapies.

8.
Diabetes ; 65(1): 34-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26470787

ABSTRACT

Nutrient stimulation of enteroendocrine L cells induces the release of the incretin and satiating peptide glucagon-like peptide 1 (GLP-1). The vagus nerve innervates visceral organs and may contribute to the mediation of gut-derived GLP-1's effects on food intake, energy homeostasis, and glycemic control. To test the hypothesis that vagal afferent neuron (VAN) GLP-1 receptors (GLP-1Rs) are necessary for these effects of endogenous GLP-1, we established a novel bilateral nodose ganglia injection technique to deliver a lentiviral vector and to knock down VAN GLP-1Rs in male Sprague Dawley rats. We found that a full expression of VAN GLP-1Rs is not necessary for the maintenance of long-term energy balance in normal eating conditions. VAN GLP-1R knockdown (kd) did, however, increase meal size and accelerated gastric emptying. Moreover, postmeal glycemia was elevated and insulin release was blunted in GLP-1R kd rats, suggesting that VAN GLP-1Rs are physiological contributors to the neuroincretin effect after a meal. Collectively, our results highlight a crucial role for the VANs in mediating the effects of endogenous GLP-1 on food intake and glycemia and may promote the further development of GLP-1-based therapies.


Subject(s)
Blood Glucose/metabolism , Eating/genetics , Energy Metabolism/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Insulin/metabolism , Neurons, Afferent/metabolism , Nodose Ganglion/metabolism , Animals , Gastric Emptying/genetics , Gene Knockdown Techniques , Insulin Secretion , Intestines/innervation , Male , Postprandial Period , Rats , Rats, Sprague-Dawley , Vagus Nerve/metabolism
9.
Physiol Behav ; 152(Pt B): 372-80, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26048300

ABSTRACT

Nutrient stimulation of the enteroendocrine L-cells induces the release of the glucagon-like peptide-1 (GLP-1), an incretin and satiating peptide. Due to its short half-life, meal-induced GLP-1's effects on food intake and glycemia are likely to be mediated in part by a paracrine signaling mechanism near the site of release. Early and recent findings from vagus nerve lesion studies scrutinized in this review strongly support an important role of the vagus nerve in mediating GLP-1's effects. Peripheral GLP-1 or GLP-1R agonist treatment failed to elicit the full satiating effects and maintain glucose homeostasis in various lesion models. The potential mechanisms underlying the vagal GLP-1R mediated satiation and glycemic control presumably involve the activation of caudal brainstem neurons via glutamatergic signaling, which activate a vagal reflex loop or/and relay the information to higher brain centers. Recent studies also presented here, however, diminish the relevance of the vagus nerve for the pharmacological intervention of obesity and diabetes with chronic GLP-1R agonist treatments, suggesting that endogenous intestinal GLP-1 and GLP-1R agonists may activate different GLP-1R populations. Finally, lesion-based approaches are limited and new technical approaches are discussed to improve the understanding of vagal GLP-1R functions in maintaining normal energy balance and its relevance in pharmacological interventions.


Subject(s)
Eating/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose/metabolism , Vagus Nerve/metabolism , Animals , Humans
10.
J Neurosci ; 35(22): 8558-69, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041922

ABSTRACT

Neurons coexpressing neuropeptide Y, agouti-related peptide, and GABA (NAG) play an important role in ingestive behavior and are located in the arcuate nucleus of the hypothalamus. NAG neurons receive both GABAergic and glutamatergic synaptic inputs, however, the developmental time course of synaptic input organization of NAG neurons in mice is unknown. In this study, we show that these neurons have low numbers of GABAergic synapses and that GABA is inhibitory to NAG neurons during early postnatal period. In contrast, glutamatergic inputs onto NAG neurons are relatively abundant by P13 and are comparatively similar to the levels observed in the adult. As mice reach adulthood (9-10 weeks), GABAergic tone onto NAG neurons increases. At this age, NAG neurons received similar numbers of inhibitory and EPSCs. To further differentiate age-associated changes in synaptic distribution, 17- to 18-week-old lean and diet-induced obesity (DIO) mice were studied. Surprisingly, NAG neurons from lean adult mice exhibit a reduction in the GABAergic synapses compared with younger adults. Conversely, DIO mice display reductions in the number of GABAergic and glutamatergic inputs onto NAG neurons. Based on these experiments, we propose that synaptic distribution in NAG neurons is continuously restructuring throughout development to accommodate the animals' energy requirements.


Subject(s)
Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/growth & development , Neurons/physiology , Synapses/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Animals , Animals, Newborn , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Synapses/genetics , Tetrodotoxin/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/pharmacology
11.
Asian-Australas J Anim Sci ; 28(4): 530-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25656200

ABSTRACT

The objective of this study was to evaluate the in vitro effects of flavonoid-rich plant extracts (PE) on ruminal fermentation characteristics and methane emission by studying their effectiveness for methanogenesis in the rumen. A fistulated Holstein cow was used as a donor of rumen fluid. The PE (Punica granatum, Betula schmidtii, Ginkgo biloba, Camellia japonica, and Cudrania tricuspidata) known to have high concentrations of flavonoid were added to an in vitro fermentation incubated with rumen fluid. Total gas production and microbial growth with all PE was higher than that of the control at 24 h incubation, while the methane emission was significantly lower (p<0.05) than that of the control. The decrease in methane accumulation relative to the control was 47.6%, 39.6%, 46.7%, 47.9%, and 48.8% for Punica, Betula, Ginkgo, Camellia, and Cudrania treatments, respectively. Ciliate populations were reduced by more than 60% in flavonoid-rich PE treatments. The Fibrobacter succinogenes diversity in all added flavonoid-rich PE was shown to increase, while the Ruminoccocus albus and R. flavefaciens populations in all PE decreased as compared with the control. In particular, the F. succinogenes community with the addition of Birch extract increased to a greater extent than that of others. In conclusion, the results of this study showed that flavonoid-rich PE decreased ruminal methane emission without adversely affecting ruminal fermentation characteristics in vitro in 24 h incubation time, suggesting that the flavonoid-rich PE have potential possibility as bio-active regulator for ruminants.

12.
J Neurosci ; 33(38): 15306-17, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048859

ABSTRACT

Neuropeptide Y (NPY) neurons in both the arcuate nucleus of the hypothalamus (ARH) and the dorsomedial hypothalamus (DMH) have been implicated in food intake and obesity. However, while ARH NPY is highly expressed in the lean animal, DMH NPY mRNA expression is observed only after diet-induced obesity (DIO). Furthermore, while ARH NPY neurons are inhibited by leptin, the effect of this adipokine on DMH NPY neurons is unknown. In this study we show that in contrast to the consistent expression in the ARH, DMH NPY mRNA expression was undetectable until after 10 weeks in mice fed a high-fat diet, and peaked at 20 weeks. Surprisingly, electrophysiological experiments demonstrated that leptin directly depolarized and increased the firing rate of DMH NPY neurons in DIO mice. To further differentiate the regulation of DMH and ARH NPY populations, fasting decreased expression of DMH NPY expression, while it increased ARH NPY expression. However, treatment with a leptin receptor antagonist failed to alter DMH NPY expression, indicating that leptin may not be the critical factor regulating mRNA expression. Importantly, we also demonstrated that DMH NPY neurons coexpress cocaine amphetamine-regulated transcript (CART); however, CART mRNA expression in the DMH peaked earlier in the progression of DIO. This study demonstrates novel and important findings. First, NPY and CART are coexpressed in the same neurons within the DMH, and second, leptin stimulates DMH NPY neurons. These studies suggest that during the progression of DIO, there is an unknown signal that drives the expression of the orexigenic NPY signal within the DMH, and that the chronic hyperleptinemia increases the activity of these NPY/CART neurons.


Subject(s)
Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Leptin/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Obesity/pathology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Diet/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hypothalamus/pathology , In Vitro Techniques , Insulin/blood , Leptin/antagonists & inhibitors , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neuropeptide Y/genetics , Obesity/blood , Obesity/etiology , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Radioimmunoassay , STAT3 Transcription Factor/metabolism , Time Factors
13.
J Comp Neurol ; 521(8): 1891-914, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23172177

ABSTRACT

The dorsomedial hypothalamus (DMH) has long been implicated in feeding behavior and thermogenesis. The DMH contains orexigenic neuropeptide Y (NPY) neurons, but the role of these neurons in the control of energy homeostasis is not well understood. NPY expression in the DMH is low under normal conditions in adult rodents but is significantly increased during chronic hyperphagic conditions such as lactation and diet-induced obesity (DIO). To understand better the role of DMH-NPY neurons, we characterized the efferent projections of DMH-NPY neurons using the anterograde tracer biotinylated dextran amine (BDA) in lactating rats and DIO mice. In both models, BDA- and NPY-colabeled fibers were limited mainly to the hypothalamus, including the paraventricular nucleus of the hypothalamus (PVH), lateral hypothalamus/perifornical area (LH/PFA), and anteroventral periventricular nucleus (AVPV). Specifically in lactating rats, BDA-and NPY-colabeled axonal swellings were in close apposition to cocaine- and amphetamine-regulated transcript (CART)-expressing neurons in the PVH and AVPV. Although the DMH neurons project to the rostral raphe pallidus (rRPa), these projections did not contain NPY immunoreactivity in either the lactating rat or the DIO mouse. Instead, the majority of BDA-labeled fibers in the rRPa were orexin positive. Furthermore, DMH-NPY projections were not observed within the nucleus of the solitary tract (NTS), another brainstem site critical for the regulation of sympathetic outflow. The present data suggest that NPY expression in the DMH during chronic hyperphagic conditions plays important roles in feeding behavior and thermogenesis by modulating neuronal functions within the hypothalamus, but not in the brainstem.


Subject(s)
Efferent Pathways/metabolism , Hyperphagia/pathology , Hypothalamus/cytology , Neurons/metabolism , Neuropeptide Y/metabolism , Obesity/pathology , Age Factors , Animals , Animals, Newborn , Biotin/analogs & derivatives , Chronic Disease , Dextrans , Disease Models, Animal , Efferent Pathways/physiology , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lactic Acid/metabolism , Male , Melanins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Obesity/etiology , Orexins , Peptide Fragments/metabolism , Pituitary Hormones/metabolism , Pregnancy , Rats , Rats, Wistar , Tryptophan Hydroxylase/metabolism
14.
Anaerobe ; 14(1): 19-28, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17996466

ABSTRACT

Responses of the rumen anaerobic fungus, Piromyces communis M014, to octadecanic long-chain fatty acids (LCFAs) were evaluated by measuring total and hydrogen gas productions, filter paper (FP) cellulose degradation and polysaccharidase enzyme activities. Octadecanic acids (stearic acid, C(18:0); oleic acid, C(18:1); linoleic acid, C(18:2) and linolenic acid, C(18:3)) were emulsified by ultrasonication under anaerobic conditions, and added to the medium at the level of 0.001%. When P. communis M014 was grown in culture with stearic and oleic acids, the cumulative gas production, FP cellulose digestion and enzyme activities were significantly (p<0.05) increased in the early incubation times relative to those for the control. However, the addition of linolenic acid inhibited all of the investigated parameters, including cellulose degradation, enzyme activities and gas production, up to 168h incubation. These results indicated that stearic and oleic acids tended to have stimulatory effects on fungal cellulolysis, whereas linolenic acid caused a significant (p<0.05) inhibitory effect on cellulolysis by the rumen fungus. The fungus, P. communis M014, can biohydrogenate C(18) unsaturated fatty acids to escape from their toxic effects. Therefore, in this study, the results indicated that the more highly the added C(18) LCFA to the fungal culture was unsaturated, the higher the inhibition of gas production and cellulase enzyme activity was.


Subject(s)
Cellulose/metabolism , Hydrogen/metabolism , Piromyces/metabolism , Rumen/microbiology , Stearic Acids/pharmacology , Ammonia/metabolism , Anaerobiosis , Animals , Cellulase/metabolism , Fermentation , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Linoleic Acid/pharmacology , Oleic Acid/pharmacology , Piromyces/drug effects , Piromyces/growth & development , Xylosidases/metabolism , alpha-Linolenic Acid/pharmacology
15.
J Pharm Sci ; 95(5): 985-97, 2006 May.
Article in English | MEDLINE | ID: mdl-16544323

ABSTRACT

Pharmacokinetics and therapeutic effects of oltipraz were evaluated after consecutive (once per day at 30 mg/kg/day for 7 and 14 days) or intermittent (once per week at 100 mg/kg/week for 1-3 weeks) oral administration to rats with liver cirrhosis induced by dimethylnitrosamine. The AUC of oltipraz was significantly greater in cirrhotic rats than controls (890 compared with 270 microg . min/mL) due to impaired liver function in cirrhotic rats. However, the AUC values after consecutive 7 (421 compared with 753 microg . min/mL) and 14 (309 compared with 821 microg . min/mL) days oral administration of oltipraz in cirrhotic rats were significantly smaller than those in respective vehicle-treated cirrhotic rats. Moreover, the AUC values after intermittent 2 and 3 weeks in cirrhotic rats were also significantly smaller than that in 1 week vehicle-treated cirrhotic rats (2370 and 1690 compared with 4760 microg . min/mL). This could be due to induction of CYP isozymes and considerably greater numbers of normal liver cells in cirrhotic rats by oral administration of oltipraz. Improved liver function by oltipraz in cirrhotic rats was proved by liver microscopy; livers are free of significant fibrosis, although evidence of bridging necrosis is still present in many rats.


Subject(s)
Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Schistosomicides/pharmacokinetics , Schistosomicides/therapeutic use , Administration, Oral , Alkylating Agents , Animals , Area Under Curve , Chromatography, High Pressure Liquid , Creatinine/metabolism , Dimethylnitrosamine , Half-Life , Hematocrit , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Organ Size/drug effects , Pyrazines/toxicity , Rats , Rats, Wistar , Schistosomicides/toxicity , Thiones , Thiophenes
16.
Int J Pharm ; 298(1): 38-46, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15890479

ABSTRACT

In order to find whether torasemide is metabolized via CYP isozymes in rats, torasemide at a dose of 2mg/kg was infused in rats pretreated with SKF 525-A, a non-specific CYP isozyme inhibitor in male Sprague-Dawley rats. The total area under the plasma concentration-time curve from time zero to time infinity (AUC) of torasemide was significantly greater in rats pretreated with SKF 525-A (a non-specific CYP isozyme inhibitor in rats) than that in control rats (3570 versus 1350 microg min/ml). This indicated that torasemide is metabolized via CYP isozymes in rats. Hence, torasemide was infused in rats pretreated with various enzyme inducers and inhibitors to find what types of CYP isozymes are involved in the metabolism of torasemide in rats. The AUC values were not significantly different in rats pretreated with 3-methylcholanthrene, phenobarbital, isoniazid, quinine and troleandomycin (main inducers of CYP1A1/2, CYP2B1/2, and CYP2E1, and main inhibitors of CYP2D1 and CYP3A1/2 in rats, respectively) compared with those in respective control rats. However, in rats pretreated with dexamethasone (a main inducer of CYP3A1/2 in rats), the AUC was significantly smaller than that in control rats (1290 versus 1590 microg min/ml). Dexamethasone probably also induces rat CYP2C11; this could be due to an increase in CYP2C11 in rats pretreated with dexamethasone. It has been reported from our laboratories that in rats pretreated with sulfaphenazole (a main inhibitor of CYP2C11 in rats) the AUC was significantly greater than that in control rats (2970 versus 1610 microg min/ml). The above data suggested that torasemide could be metabolized in male rats mainly via CYP2C11.


Subject(s)
Sulfonamides/pharmacokinetics , Animals , Aryl Hydrocarbon Hydroxylases/physiology , Cytochrome P-450 CYP3A , Cytochrome P450 Family 2 , Dexamethasone/pharmacology , Enzyme Induction , Male , Membrane Proteins/physiology , Rats , Rats, Sprague-Dawley , Steroid 16-alpha-Hydroxylase/physiology , Sulfonamides/administration & dosage , Torsemide
17.
Eur J Pharm Sci ; 25(2-3): 337-45, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15911231

ABSTRACT

Effects of diabetes mellitus induced by streptozotocin (DMIS) on the pharmacokinetics of DA-7867 were investigated after i.v. and oral administration (10mg/kg) to control Sprague-Dawley rats and DMIS rats (at 7th and 29th days after administration of streptozotocin, 45mg/kg). After i.v. administration to DMIS rats, the AUC(0-infinity) values were significantly smaller (50.7 and 64.8% decrease for 7th and 29th days, respectively); this could be due to significantly faster Cl values in the rats (127 and 183% increase for 7th and 29th days, respectively). The faster Cl values were mainly due to significantly greater amount of unchanged drug excreted in 24-h urine (Ae(0-24h)). The greater Ae(0-24h) in DMIS rats could be due to urine flow rate-dependent renal clearance of DA-7867. After oral administration to DMIS rats, the AUC(0-infinity) values were also significantly smaller (61.3 and 72.6% decrease for 7th and 29th days, respectively); this could also be mainly due to significantly greater Ae(0-24h) in the rats. Streptozotocin-induced hepatotoxicity did not influence considerably on the pharmacokinetics of DA-7867 at 7th day when compared with those at 29th day.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Oxazolidinones/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Injections, Intravenous , Male , Metabolic Clearance Rate , Oxazolidinones/administration & dosage , Rats , Rats, Sprague-Dawley , Streptozocin , Time Factors
18.
Biopharm Drug Dispos ; 26(3): 99-115, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15723427

ABSTRACT

Dose-independent pharmacokinetics of oltipraz after intravenous and/or oral administration at various doses to mice, rats, rabbits and dogs were evaluated. After both intravenous and/or oral administration of oltipraz to mice (5, 10 and 20 mg/kg for intravenous and 15, 30 and 50 mg/kg for oral administration), rats (5, 10 and 20 mg/kg for intravenous and 25, 50 and 100 mg/kg for oral administration), rabbits (5, 10 and 30 mg/kg for intravenous administration) and dogs (5 and 10 mg/kg for intravenous and 50 and 100 mg/kg for oral administration), the total area under the plasma concentration-time curve from time zero to time infinity (AUC) values of oltipraz were dose-proportional in all animals studied. Animal scale-up of some pharmacokinetics parameters of oltipraz was also performed based on the parameters after intravenous administration at a dose of 10 mg/kg to mice, rats, rabbits and dogs. Linear relationships were obtained between log time-averaged total body clearance (Cl) x maximum life-span potential (MLP) (1 year/h) and log species body weight (W) (kg) (r=0.999; p=0.0015), log Cl (l/h) and log W (kg) (r=0.979; p=0.0209), and log apparent volume of distribution at steady state (V(ss)) (l) and log W (kg) (r=0.999; p=0.0009). The corresponding allometric equations were ClxMLP=49.8 W(0.861), Cl=5.20 W(0.523) and V(ss)=4.46 W(0.764). Interspecies scale-up of plasma concentration-time data for the four species using pharmacokinetic time of dienetichron resulted in similar profiles. In addition, concentrations of oltipraz in a plasma concentration-time profile for humans predicted using the four animal data fitted to the dienetichron time transformation of animal data.


Subject(s)
Pyrazines/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Dogs , Half-Life , Humans , Injections, Intravenous , Metabolic Clearance Rate , Mice , Predictive Value of Tests , Pyrazines/administration & dosage , Pyrazines/blood , Rabbits , Rats , Species Specificity , Thiones , Thiophenes , Tissue Distribution
19.
Biopharm Drug Dispos ; 26(2): 77-83, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15617135

ABSTRACT

Pharmacokinetic parameters of oltipraz were compared after intravenous and oral administration at a dose of 30 mg/kg to control rats and rats with water deprivation for 72 h (rats with dehydration). The plasma protein binding of oltipraz was measured in both groups of rats using an equilibrium dialysis technique. The concentrations of oltipraz were measured by the reported HPLC analysis. After intravenous administration, the total area under the plasma concentration-time curve from time zero to time infinity (AUC), terminal half-life, time-averaged total body and nonrenal clearances, and apparent volume of distribution at steady state were not significantly different between the two groups of rats. However, after oral administration to rats with dehydration, the AUC was significantly smaller than that in control rats (180 versus 316 microg min/ml) mainly due to decrease in absorption. In rats with dehydration, plasma protein binding was significantly greater than that in control rats (91.5 +/- 0.309 versus 81.3 +/- 2.79%).


Subject(s)
Anticarcinogenic Agents/pharmacokinetics , Pyrazines/pharmacokinetics , Water Deprivation/physiology , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/metabolism , Area Under Curve , Blood Proteins/metabolism , Gastrointestinal Tract/metabolism , Injections, Intravenous , Intestinal Absorption , Male , Metabolic Clearance Rate , Protein Binding , Pyrazines/administration & dosage , Pyrazines/blood , Rats , Rats, Sprague-Dawley , Thiones , Thiophenes , Time Factors
20.
J Pharm Sci ; 93(9): 2353-63, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15295795

ABSTRACT

Pharmacokinetic parameters of oltipraz were compared after intravenous and oral administration at a dose of 30 mg/kg to control rats and rats with U-ARF. After intravenous administration to rats with U-ARF, the AUC was significantly greater (1100 versus 1730 microg x min/mL) than that in control rats, and this could be due to significantly slower CL (27.2 versus 17.3 mL/min/kg). The slower CL could be mainly due to significantly slower CL(NR) (27.2 versus 17.3 mL/min/kg), and this could be supported by significantly slower in vitro CL(int) (32.1 versus 13.2 mL/min/whole liver) in the rats. The Vss was significantly larger in rats with U-ARF (4050 versus 5680 mL/kg), and this was not due to a significant increase in free fractions (unbound in plasma proteins) of oltipraz in the rats because the free fractions were 17.0 and 15.7% for control rats and rats with U-ARF, respectively. Unexpectedly, after oral administration to rats with U-ARF, the AUC of oltipraz was significantly smaller than that in control rats (329 versus 149 microg x min/mL), and this could be mainly due to a decrease in the absorption of oltipraz from the gastrointestinal tract in the rats (95 and 72% of the oral dose were absorbed in control rats and rats with U-ARF, respectively).


Subject(s)
Acute Kidney Injury/blood , Pyrazines/blood , Pyrazines/pharmacokinetics , Animals , Male , Pyrazines/administration & dosage , Rats , Rats, Sprague-Dawley , Thiones , Thiophenes
SELECTION OF CITATIONS
SEARCH DETAIL
...