Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 483
Filter
1.
Oncogene ; 36(15): 2054-2065, 2017 04.
Article in English | MEDLINE | ID: mdl-27721405

ABSTRACT

Androgens are known to protect prostate cancer cells from DNA damage. Recent studies showed regulation of DNA repair genes by androgen receptor signaling in prostate cancers. ELL-associated factor 2 (EAF2) is an androgen-regulated tumor suppressor and its intracellular localization can be modulated by ultraviolet light, suggesting a potential role for EAF2 in androgen regulation of DNA repair in prostate cancer cells. Here we show that knockdown of EAF2 or its homolog EAF1 sensitized prostate cancer cells to DNA damage and the sensitization did not require p53. EAF2 knockout mouse prostate was also sensitized to γ-irradiation. Furthermore, EAF2 knockdown blocked androgen repression of LNCaP or C4-2 cells from doxorubicin induction of γH2ax, a DNA damage marker. In human prostate cancer specimens, EAF2 expression was inversely correlated with the level of γH2ax. Further analysis showed that EAF2 and EAF1 are required for the recruitment and retention of Ku70/Ku80 to DNA damage sites and play a functional role in nonhomologous end-joining DNA repair. These findings provide evidence for EAF2 as a key factor mediating androgen protection of DNA damage via Ku70/Ku80 in prostate cancer cells.


Subject(s)
DNA Damage , DNA End-Joining Repair , Ku Autoantigen/metabolism , Prostatic Neoplasms/genetics , Transcription Factors/genetics , Androgens/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , Humans , Ku Autoantigen/genetics , Male , Mice, Inbred C57BL , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/metabolism
2.
J Neuroendocrinol ; 28(4)2016 04.
Article in English | MEDLINE | ID: mdl-27114001

ABSTRACT

Central oxytocin (OT) promotes feeding termination in response to homeostatic challenges, such as excessive stomach distension, salt loading and toxicity. OT has also been proposed to affect feeding reward by decreasing the consumption of palatable carbohydrates and sweet tastants. Because the OT receptor (OTR) is expressed in the nucleus accumbens core (AcbC) and shell (AcbSh), a site regulating diverse aspects of eating behaviour, we investigated whether OT acts there to affect appetite in rats. First, we examined whether direct AcbC and AcbSh OT injections affect hunger- and palatability-driven consumption. We found that only AcbC OT infusions decrease deprivation-induced chow intake and reduce the consumption of palatable sucrose and saccharin solutions in nondeprived animals. These effects were abolished by pretreatment with an OTR antagonist, L-368,899, injected in the same site. AcbC OT at an anorexigenic dose did not induce a conditioned taste aversion, which indicates that AcbC OT-driven anorexia is not caused by sickness/malaise. The appetite-specific effect of AcbC OT is supported by the real-time polymerase chain reaction analysis of OTR mRNA in the AcbC, which revealed that food deprivation elevates OTR mRNA expression, whereas saccharin solution intake decreases OTR transcript levels. We also used c-Fos immunohistochemistry as a marker of neuronal activation and found that AcbC OT injection increases activation of the AcbC itself, as well as of two feeding-related sites: the hypothalamic paraventricular and supraoptic nuclei. Finally, considering the fact that OT plays a significant role in social behaviour, we examined whether offering animals a meal in a social setting would modify their hypophagic response to AcbC OT injections. We found that a social context abolishes the anorexigenic effects of AcbC OT. We conclude that OT acting via the AcbC decreases food intake driven by hunger and reward in rats offered a meal in a nonsocial setting.


Subject(s)
Eating/physiology , Nucleus Accumbens/physiology , Oxytocin/physiology , Animals , Appetite , Camphanes/pharmacology , Feeding Behavior/physiology , Food Deprivation/physiology , Male , Microinjections , Neurons/physiology , Oxytocin/administration & dosage , Oxytocin/antagonists & inhibitors , Oxytocin/biosynthesis , Paraventricular Hypothalamic Nucleus/physiology , Piperazines/pharmacology , Rats , Social Behavior , Supraoptic Nucleus/physiology
3.
J Neuroendocrinol ; 28(4)2016 04.
Article in English | MEDLINE | ID: mdl-26918919

ABSTRACT

Central oxytocin suppresses appetite. Neuronal activity and the release of oxytocin coincide with satiation, as well as with adverse events (e.g. hyperosmolality, toxicity or excessive stomach distension) that necessitate an immediate termination of eating behaviour. Oxytocin also decreases consumption driven by reward, especially as derived from ingesting carbohydrates and sweet tastants. This review summarises current knowledge of the role of oxytocin in food intake regulation and highlights a growing body of evidence showing that oxytocin is a conditional anorexigen [i.e. its effects on appetite differ significantly with respect to certain (patho)physiological, behavioural and social contexts].


Subject(s)
Anorexia/chemically induced , Anorexia/psychology , Appetite Regulation/physiology , Appetite/physiology , Oxytocin/physiology , Animals , Humans , Social Environment
4.
Pharmacol Biochem Behav ; 100(3): 575-80, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21925202

ABSTRACT

Butorphanol ([BT] an opioid receptor agonist/antagonist) is different from other opioid agonists in that a single dose of BT can elicit up to 12 g of chow intake in a satiated rat whereas most opioid agonists induce a mild feeding response (2-3 g). Here, we first examined whether the effectiveness of BT to elicit feeding was affected by dose, method of infusion and possible tachyphylaxis following administration. Secondly, we examined whether BT administration influenced hypothalamic NPY gene expression and peptide levels. A single dose administration of BT (4 mg/kg) significantly increased food intake at 2, 3 and 6 h after administration. However following repeated injections of BT at 4 mg/kg, the cumulative long-term intake of BT-treated rats did not differ from that of controls, indicating that the animals compensate for the increased feeding following BT injection by decreased feeding at a later time. An ascending dose schedule of repeated BT injections resulted in additional feeding. NPY gene expression in the ARC was influenced by how much food had been consumed, but not by BT. The amount of food consumed and the level of NPY mRNA were inversely correlated. This is consistent with NPY's role in normal feeding. BT treatment did not affect either NPY or leptin RIA levels. We conclude that the feeding produced by BT is sensitive to dose and dosing paradigm. Further, its mechanism of action does not appear to be mediated by NPY or leptin pathways.


Subject(s)
Analgesics, Opioid/pharmacology , Appetite Stimulants/pharmacology , Arcuate Nucleus of Hypothalamus/drug effects , Butorphanol/pharmacology , Energy Intake/drug effects , Narcotic Antagonists/pharmacology , Neuropeptide Y/metabolism , Analgesics, Opioid/administration & dosage , Animals , Appetite Stimulants/administration & dosage , Arcuate Nucleus of Hypothalamus/metabolism , Behavior, Animal/drug effects , Butorphanol/administration & dosage , Dose-Response Relationship, Drug , Food Deprivation , Gene Expression Regulation/drug effects , Male , Narcotic Antagonists/administration & dosage , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/genetics , Organ Specificity , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Tachyphylaxis
5.
Neuroscience ; 171(3): 779-87, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20875839

ABSTRACT

The nucleus accumbens (NAcc) mediates feeding reward; its activity reflects tastants' hedonic value. NAcc dopamine guides immediate responses to reward, however, its involvement in establishing long-term responses after a period of exposure to palatable foods has not been defined. Furthermore, reward-driven overeating propels weight increase, but the scale of weight gain depends on animals' obesity-prone (OP) or -resistant (OR) phenotype. It is unclear whether the NAcc dopamine response to palatable food depends on obesity susceptibility. We investigated the effect of unrestricted extended access to high-fat high-sugar (HFHS) diet on expression of genes encoding dopamine receptors in the NAcc of OP and OR rats. We examined persistence of HFHS diet-induced changes in D(1) and D(2) gene expression in OP and OR rats subjected to HFHS withdrawal (bland chow for 18 days). Effects of restricted access to HFHS by pair-feeding were also studied. Using reverse transcriptase PCR (RT-PCR), we found that NAcc D(1) mRNA was downregulated after long-term HFHS access in OP vs. OR animals. The effect was also observed after 18 days of HFHS withdrawal. Furthermore, restricted HFHS led to downregulation of D(1) as well as of D(2) mRNA levels compared to chow-fed controls. A difference in the expression of mu opioid receptor in the NAcc was also detected between the OP and OR rats during access to palatable food but not after withdrawal. We conclude that exposure to HFHS diets has lasting consequences for the NAcc dopamine system, perhaps modifying the motivation to search for food reward. The fact that the NAcc D(1) expression changes in OP animals after long-term exposure to palatable food and that this effect extends well into the reward discontinuation phase, implicates the D(1) receptor in the propensity to overeat and, in effect, gain weight in obesity prone individuals.


Subject(s)
Appetite Regulation/genetics , Down-Regulation/genetics , Eating/genetics , Nucleus Accumbens/metabolism , Obesity/genetics , Phenotype , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/genetics , Animals , Disease Models, Animal , Down-Regulation/physiology , Male , Nucleus Accumbens/physiopathology , Obesity/metabolism , Obesity/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/physiology , Time Factors , Weight Gain/genetics
6.
Int J Obes (Lond) ; 33 Suppl 2: S54-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528981

ABSTRACT

Humans eat for many reasons, including the rewarding qualities of foods. A host of neurotransmitters have been shown to influence eating behavior and some of these appear to be involved in reward-induced eating. Endogenous opioid peptides and their receptors were first reported more than 30 years ago, and studies suggesting a role of opioids in the regulation of food intake date back nearly as far. Opioid agonists and antagonists have corresponding stimulatory and inhibitory effects on feeding. In addition to studies aimed at identifying the relevant receptor subtypes and sites of action within the brain, there has been a continuing interest in the role of opioids on diet/taste preferences, food reward, and the overlap of food reward with others types of reward. Data exist that suggest a role for opioids in the control of appetite for specific macronutrients, but there is also evidence for their role in the stimulation of intake based on already-existing diet or taste preferences and in controlling intake motivated by hedonics rather than by energy needs. Finally, various types of studies indicate an overlap between mechanisms mediating drug reward and palatable food reward. Preference or consumption of sweet substances often parallels the self-administration of several drugs of abuse, and under certain conditions, the termination of intermittent access to sweet substances produces symptoms that resemble those observed during opiate withdrawal. The overconsumption of readily available and highly palatable foods likely contributes to the growing rates of obesity worldwide. An understanding of the role of opioids in mediating food reward and promoting the overconsumption of palatable foods may provide insights into new approaches for preventing obesity.


Subject(s)
Appetite Regulation/drug effects , Appetite Regulation/physiology , Feeding Behavior/physiology , Obesity/physiopathology , Opioid Peptides/physiology , Receptors, Opioid/physiology , Reward , Food , Humans , Narcotic Antagonists/pharmacology , Neural Pathways/drug effects , Neural Pathways/physiology , Obesity/psychology , Opioid Peptides/antagonists & inhibitors , Taste/physiology
7.
Genes Brain Behav ; 8(2): 193-202, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19077174

ABSTRACT

The aim of this study was to investigate whether the preference for a palatable high-fat diet (HFD) is associated with response to novelty and with anxiety-like behavior in rats and whether such fat preference correlates with gene expression of hypothalamic neuropeptides related to feeding. We subjected male rats to two tests of exploration of novel environments: the multivariate concentric square field (MCSF) and the elevated plus maze (EPM). The rats were then exposed to a 5-day test of preference for a palatable HFD versus reference diets. Messenger RNA (mRNA) levels of 21 neuropeptides were investigated by quantitative polymerase chain reaction. We found a strong positive correlation of HFD preference and open-arm activity in the EPM (% open-arm time, r(s) = 0.629, df = 26, P < 0.001). Thus, HFD preference was inversely associated with anxiety-like behavior. The same association was found for HFD preference and behavior in the MCSF (bridge entries, r(s) = 0.399, df = 23, P = 0.048). In addition, the HFD preference was positively correlated (r(s) = 0.433, df = 25, P = 0.021) with hypothalamic mRNA levels of urocortin 2 (Ucn 2). Moreover, behavior in the EPM was significantly correlated with expression levels of the receptor for Ucn 2, the corticotropin-releasing factor receptor 2, in the hypothalamus (r(s) = 0.382, df = 33, P = 0.022, pituitary (r(s) = 0.494, df = 31, P = 0.004) and amygdala (r(s) = 0.381, df = 30, P = 0.032). We conclude that preference for palatable HFD is inversely associated with anxiety and propose that Ucn 2 signaling may play a role in this association.


Subject(s)
Anxiety/psychology , Dietary Fats , Food Preferences/physiology , Urocortins/physiology , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Eating/genetics , Eating/physiology , Emotions/physiology , Exploratory Behavior/physiology , Gene Expression , Hormones/blood , Hypothalamus/metabolism , Individuality , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Urocortins/genetics , Weight Gain/genetics , Weight Gain/physiology
8.
Am J Physiol Regul Integr Comp Physiol ; 291(4): R889-99, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16763079

ABSTRACT

Selectively-bred obesity-resistant [diet resistant (DR)] rats weigh less than obesity-prone [diet-induced obese (DIO)] rats, despite comparable daily caloric intake, suggesting phenotypic energy expenditure differences. Human data suggest that obesity is maintained by reduced ambulatory or spontaneous physical activity (SPA). The neuropeptide orexin A robustly stimulates SPA. We hypothesized that DR rats have greater: 1) basal SPA, 2) orexin A-induced SPA, and 3) preproorexin, orexin 1 and 2 receptor (OX1R and OX2R) mRNA, compared with DIO rats. A group of age-matched out-bred Sprague-Dawley rats were used as additional controls for the behavioral studies. DIO, DR, and Sprague-Dawley rats with dorsal-rostral lateral hypothalamic (rLHa) cannulas were injected with orexin A (0, 31.25, 62.5, 125, 250, and 500 pmol/0.5 microl). SPA and food intake were measured for 2 h after injection. Preproorexin, OX1R and OX2R mRNA in the rLHa, and whole hypothalamus were measured by real-time RT-PCR. Orexin A significantly stimulated feeding in all rats. Orexin A-induced SPA was significantly greater in DR and Sprague-Dawley rats than in DIO rats. Two-mo-old DR rats had significantly greater rLHa OX1R and OX2R mRNA than DIO rats but comparable preproorexin levels. Eight-mo-old DR rats had elevated OX1R and OX2R mRNA compared with DIO rats, although this increase was significant for OX2R only at this age. Thus DR rats show elevated basal and orexin A-induced SPA associated with increased OX1R and OX2R gene expression, suggesting that differences in orexin A signaling through OX1R and OX2R may mediate DIO and DR phenotypes.


Subject(s)
Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Obesity/metabolism , Obesity/physiopathology , Receptors, Neuropeptide/metabolism , Age Factors , Animals , Energy Intake/drug effects , Energy Intake/physiology , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Motor Activity/physiology , Neuropeptides/pharmacology , Orexin Receptors , Orexins , Phenotype , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/genetics , Signal Transduction/physiology , Species Specificity
9.
Psychopharmacology (Berl) ; 182(1): 75-83, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16075284

ABSTRACT

RATIONALE: Centrally administered orexin A induces both feeding and locomotion in rats. Thus, the feeding response following orexin A administration may be secondary to general increases in activity rather than a specific motivation to eat. OBJECTIVE: The aim of the study is to determine whether orexin A increases the motivation to eat. METHODS: The effect of orexin A (0, 31.25, 62.5, 125, 250, and 500 pmol) on breakpoint was determined in male Sprague-Dawley rats with rostro-lateral hypothalamic cannulae under a progressive ratio of five schedule (PR5). The effect of orexin A (0, 31.25, 125, and 500 pmol) on pressing rate under a fixed ratio (20) schedule was obtained to analyze the time course of orexin-A-induced pressing. The effect of 24-h food deprivation on breakpoint under PR5 and the effect of orexin A (125 pmol) on free feeding (sweet pellets) and on open-field locomotor activity (0, 100, 500, and 1,000 pmol) were also tested. RESULTS: Orexin A significantly augmented free feeding of sweet pellets, open-field locomotor activity, rate of pressing (FR20 schedule), and breakpoint (PR5 schedule), although compared to 24-h deprivation, the effect of orexin A on breakpoint was mild. However, there was a differential dose response relationship and time course of stimulation between orexin A's effects on locomotion and lever pressing. CONCLUSION: These data indicate that infusion of orexin A enhances free feeding by enhancing and possibly prolonging motivation to eat.


Subject(s)
Appetite/drug effects , Feeding Behavior/drug effects , Hypothalamic Area, Lateral/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Taste/drug effects , Animals , Appetitive Behavior/drug effects , Dose-Response Relationship, Drug , Motor Activity/drug effects , Nerve Net/drug effects , Orexins , Rats , Rats, Sprague-Dawley , Reinforcement Schedule
10.
Behav Neurosci ; 118(1): 173-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14979794

ABSTRACT

Rats were trained to respond under a cyclic-ratio schedule of reinforcement composed of an ascending, followed by a descending, sequence of ratio values. Subjects were trained while exposed to 70 dB white noise, then tested while exposed to 70 or 90 dB white noise. Exposure to 90 dB white noise elevated the response function (p<.02). Naloxone was then administered intraperitoneally at 0.3. 1.0. and 3.0 mg/kg under 70 dB and 90 dB white noise. Naloxone administration (1.0 and 3.0 mg/kg) significantly depressed the response function obtained under 90 dB white noise (ps<.01) but did not affect the function obtained under 70 dB white noise. These findings suggest that mild stress increases food intake through a mechanism affecting palatability enhanced by the release of endogenous opioids.


Subject(s)
Feeding Behavior/physiology , Reaction Time/physiology , Reinforcement Schedule , Stress, Physiological/metabolism , Acoustic Stimulation/methods , Animals , Feeding Behavior/drug effects , Male , Naloxone/pharmacology , Narcotics/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects
11.
Am J Physiol Regul Integr Comp Physiol ; 281(5): R1605-12, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641133

ABSTRACT

The opioid receptor antagonist naloxone decreases consumption of high-sucrose diets but does not reduce cornstarch diet intake in energy-restricted rats. Sucrose-fed rats eat at a much higher rate, consuming more food than cornstarch-fed rats. We examined meal microstructure using an automated weighing system in food-restricted rats eating either a high-sucrose or high-cornstarch diet. Sucrose-fed rats exhibited a higher rate of eating during their first meal compared with cornstarch-fed rats (0.34 vs. 0.20 g/min, respectively). However, naloxone did not reduce eating rate in either group. Naloxone decreased the size of the first meal in both diet groups by shortening the length of the meal. Naloxone's anorectic effect was more potent in the sucrose-fed rats. These results indicate that naloxone's heightened anorectic effect on sucrose diet consumption is not "rate dependent." Naloxone's anorectic actions may be modulated by two conditions, the sensory properties of food and the energy state of the animal. Thus the elevated anorectic potency of naloxone in energy-restricted sucrose-fed rats may reflect actions on neural systems that mediate orosensory and/or postingestive signals.


Subject(s)
Dietary Sucrose/administration & dosage , Eating/drug effects , Naloxone/pharmacology , Starch/administration & dosage , Animals , Diet , Eating/physiology , Feeding Behavior/physiology , Male , Rats , Rats, Sprague-Dawley , Time Factors
12.
Brain Res ; 909(1-2): 75-80, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11478923

ABSTRACT

Antagonists selective for either kappa- [e.g. nor-binaltorphimine (nor-BNI)] and mu- (e.g. beta-funaltrexamine) opioid receptors have previously been shown to reduce both kappa- and mu-opioid-induced feeding. In the present studies, the anorectic effects of GNTI, a newly synthesized antagonist selective for kappa-opioid receptors, were studied in rats. GNTI (0.032-0.32 nmol; i.c.v.), administered 15 min prior to food access, reduced feeding induced by the kappa-opioid agonist U50,488 (producing a 70% maximal decrease), the mu-opioid agonist DAMGO (90% maximal decrease), and 24 h acute food deprivation (60% maximal decrease). GNTI did not reduce the orexigenic effects of butorphanol, an agonist that binds to both kappa- and mu-opioid receptors, and neuropeptide Y (NPY). Taken together, these results suggest that GNTI is a potent anorectic agent and opioid antagonist in rats. Like nor-BNI, GNTI reduced feeding induced by both kappa- and mu-opioid agonists. However, unlike nor-BNI, GNTI did not alter the orexigenic effects of butorphanol or NPY. Given the selectivity of GNTI and its effectiveness in several of the present experiments, its potency, and its short duration of action compared to nor-BNI, GNTI may serve to be a useful tool to study behavioral effects mediated by kappa-opioid receptors.


Subject(s)
Brain/drug effects , Eating/drug effects , Food Deprivation/physiology , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Neuropeptide Y/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/pharmacology , Animals , Brain/metabolism , Butorphanol/pharmacology , Drug Interactions/physiology , Eating/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Guanidines , Male , Morphinans , Naltrexone/pharmacology , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism
13.
Am J Physiol Regul Integr Comp Physiol ; 281(2): R673-80, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11448874

ABSTRACT

Central injection of alpha-melanocyte-stimulating hormone (alpha-MSH) decreases food intake, suggesting a role for this peptide in the mediation of satiety. Inasmuch as alpha-MSH also supports the development of taste aversions under certain conditions, the nature of its influence on ingestive behavior, i.e., whether it is related to satiety or aversion, remains unclear. In the present studies, we used immunostaining, including that for c-Fos as a marker of neuronal activation, to further substantiate the physiological role for alpha-MSH in the regulation of consummatory behavior. We found that an increase in activation of alpha-MSH neurons in the arcuate nucleus coincided with meal termination. Administration of powerful aversive agents, LiCl and CuSO(4), did not stimulate alpha-MSH cells but did induce pronounced activation of oxytocin (OT) and vasopressin (VP) neurons, the final components of circuitry mediating aversion. We observed fewer Fos-positive OT/VP neurons after alpha-MSH injection into the lateral ventricle or into the hypothalamic paraventricular nucleus, treatments that cause mild or no aversion, respectively. The degree of activation of OT/VP neurons paralleled the magnitude of aversive response to a given treatment. Our data support the hypothesis that, in the arcuate nucleus, alpha-MSH acts as a satiety mediator independent from aversion-related mechanisms.


Subject(s)
Consummatory Behavior , Feeding Behavior , Neurons/chemistry , alpha-MSH/physiology , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Arcuate Nucleus of Hypothalamus/cytology , Consummatory Behavior/drug effects , Copper Sulfate/administration & dosage , Feeding Behavior/drug effects , Lithium Chloride/administration & dosage , Male , Oxytocin/analysis , Paraventricular Hypothalamic Nucleus/chemistry , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley , Supraoptic Nucleus/chemistry , Vasopressins/analysis , alpha-MSH/analysis , alpha-MSH/pharmacology
14.
Neuroreport ; 12(8): 1727-30, 2001 Jun 13.
Article in English | MEDLINE | ID: mdl-11409748

ABSTRACT

The aim of our experiments was to study the presumed functional relationship between the melanocortin and opioid systems in the regulation of food intake. We determined that a non-selective opioid receptor antagonist, naltrexone, at relatively low doses, decreases food intake induced by i.c.v. agouti-related protein (Agrp). We also observed that peripheral injection of naltrexone at a dose known to produce anorexigenic responses induced c-Fos immunoreactivity in significantly more arcuate nucleus alpha-MSH neurons than observed in control animals. The results of our study support the notion that the melanocortin and opioid systems interact in the regulation of food intake. Based on these data we speculate that opioid peptides suppress alpha-MSH-dependent satiety mechanisms; conversely, it is possible that the orexigenic action of Agrp is mediated via opioid dependent circuitry.


Subject(s)
Eating/physiology , Endorphins/physiology , alpha-MSH/physiology , Agouti-Related Protein , Animals , Brain/cytology , Brain/drug effects , Brain/physiology , Eating/drug effects , Immunohistochemistry , Injections, Intraventricular , Injections, Subcutaneous , Intercellular Signaling Peptides and Proteins , Male , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/physiology , Proteins/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
15.
Brain Res ; 907(1-2): 125-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11430893

ABSTRACT

Butorphanol (BT), a mixed kappa- and mu-opioid receptor agonist, induces vigorous food intake in rats. Peripheral injection of BT seems to increase food intake more effectively than intracerebroventricular administration. To further elucidate the nature of BT's influence on consummatory behavior, we examined which feeding-related brain areas exhibit increased c-Fos immunoreactivity (IR) following subcutaneous injection of 4 mg/kg body weight BT, a dose known to induce a maximal orexigenic response. We also evaluated whether direct administration of BT into the forebrain regions activated by peripheral BT injection affects food intake. Peripheral BT administration induced c-Fos-IR in the hypothalamic paraventricular nucleus (PVN), central nucleus of the amygdala (CeA), and nucleus of the solitary tract (NTS). However, 0.1-30 microg BT infused into the CeA, failed to increase food intake 1, 2, and 4 h after injection. Only the highest dose of BT (30 microg) injected into the PVN increased feeding. These results suggest that the PVN, CeA, and NTS mediate the effects of peripherally-injected BT. The PVN or CeA are probably not the main target sites of immediate BT action.


Subject(s)
Amygdala/drug effects , Appetite Stimulants/pharmacology , Appetite/drug effects , Butorphanol/pharmacology , Eating/drug effects , Narcotic Antagonists/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Solitary Nucleus/drug effects , Amygdala/chemistry , Amygdala/physiology , Animals , Appetite Stimulants/administration & dosage , Biomarkers , Butorphanol/administration & dosage , Caudate Nucleus/chemistry , Injections , Injections, Intraventricular , Injections, Subcutaneous , Male , Narcotic Antagonists/administration & dosage , Nerve Tissue Proteins/analysis , Nucleus Accumbens/chemistry , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/physiology , Proto-Oncogene Proteins c-fos/analysis , Putamen/chemistry , Rats , Rats, Sprague-Dawley , Septal Nuclei/chemistry , Solitary Nucleus/chemistry , Solitary Nucleus/physiology
17.
Peptides ; 22(1): 129-34, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11179607

ABSTRACT

alpha-Melanocyte-stimulating hormone (alpha-MSH) appears to play a tonic inhibitory role in feeding and energy storage. MTII, a specific synthetic MC3-R/MC4-R agonist, has similar effects on feeding in rats. The current studies demonstrate that PVN administration of alpha-MSH or MTII decreases nocturnal and NPY-stimulated food intake without causing aversive effects. Co-administration with NPY of 600 pmol alpha-MSH or 1 pmol MTII into the PVN caused a significant decrease in NPY-induced feeding. PVN administration of MTII or alpha-MSH at doses effective to suppress feeding did not cause conditioned taste aversion (CTA). ICV administration of alpha-MSH, however, did cause weak CTA. These results indicate that the potent effects on feeding of MC3-R and MC4-R agonists when injected into the PVN are not due to aversive effects.


Subject(s)
Eating/physiology , Hypothalamus/physiology , Receptors, Corticotropin/physiology , alpha-MSH/physiology , Animals , Eating/drug effects , Male , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 3 , Receptor, Melanocortin, Type 4 , Receptors, Corticotropin/agonists , alpha-MSH/administration & dosage , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
18.
Am J Physiol Regul Integr Comp Physiol ; 280(2): R473-80, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208577

ABSTRACT

Ventricular administration of urocortin (UCN) inhibits feeding, but specific site(s) of UCN action are unknown. In the current studies we examined the effect of UCN in the hypothalamic paraventricular nucleus (PVN) on feeding. We tested UCN administered into the PVN in several paradigms: deprivation-induced, nocturnal, and neuropeptide Y (NPY)-induced feeding. We compared the effect of equimolar doses of UCN and corticotrophin releasing hormone (CRH) on NPY-induced and nocturnal feeding, determined whether UCN in the PVN produced a conditioned taste aversion (CTA) and induced changes in c-Fos immunoreactivity (c-Fos-ir) after UCN and NPY administration in the PVN. UCN in the PVN significantly decreased NPY and nocturnal and deprivation-induced feeding at doses of 1, 10, and 100 pmol, respectively. UCN anorectic effects lasted longer than those attributed to CRH. Ten and thirty picomoles UCN did not induce a CTA, whereas 100 pmol UCN produced a CTA. UCN (100 pmol) in the PVN neither increased c-Fos-ir in any brain region assayed nor altered c-Fos-ir patterns resulting from PVN NPY administration. These data suggest the hypothalamic PVN as a site of UCN action.


Subject(s)
Brain/physiology , Corticotropin-Releasing Hormone/pharmacology , Feeding Behavior/drug effects , Paraventricular Hypothalamic Nucleus/physiology , Animals , Brain/drug effects , Circadian Rhythm , Corticotropin-Releasing Hormone/administration & dosage , Dose-Response Relationship, Drug , Energy Intake/drug effects , Energy Intake/physiology , Food Deprivation , Genes, fos , Male , Microinjections , Neuropeptide Y/administration & dosage , Neuropeptide Y/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Urocortins
19.
Neuroreport ; 11(14): 3251-5, 2000 Sep 28.
Article in English | MEDLINE | ID: mdl-11043558

ABSTRACT

Cocaine and amphetamine regulated transcript (CART) decreases feeding and body weight after ventricular injection. CART mRNA and peptide are found in the paraventricular nucleus of the hypothalamus (PVN). The purpose of the present study was to determine effects of PVN-injected CART on feeding and thermogenic capacity. PVN-injected CART (55-102, 100 pmol) significantly decreased NPY-induced feeding at 1, 2 and 4 h, but did not significantly affect deprivation-induced feeding. CART induced gene expression of uncoupling protein 1 (UCP1), UCP2, and UCP3 in brown and white adipose tissue and biceps femoris muscle respectively. These results indicate the PVN as a specific site of CART action, and suggest that CART in the PVN may have an important influence on energy metabolism.


Subject(s)
Carrier Proteins/genetics , Eating/drug effects , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Membrane Proteins/genetics , Membrane Transport Proteins , Mitochondrial Proteins , Nerve Tissue Proteins/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Thermogenesis/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Eating/physiology , Energy Metabolism/physiology , Fatty Acids, Nonesterified/blood , Food Deprivation/physiology , Gene Expression Regulation/physiology , Ion Channels , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , Proteins/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Thermogenesis/physiology , Uncoupling Protein 1 , Uncoupling Protein 2 , Uncoupling Protein 3
20.
Am J Physiol Regul Integr Comp Physiol ; 279(4): R1504-11, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11004021

ABSTRACT

Aversive properties of lithium chloride (LiCl) are mediated via pathways comprising neurons of the nucleus of the solitary tract (NTS) and oxytocin (OT) and vasopressin (VP) cells in the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei. Because opioids act on brain regions that mediate effects of LiCl, we evaluated whether administration of opioids shortly before LiCl in rats influences 1) development of conditioned taste aversion (CTA) and 2) activation of NTS neurons and OT/VP cells. Neuronal activation was assessed by applying c-Fos immunohistochemical staining. Three opioids were used: morphine (MOR), a mu-agonist, butorphanol tartrate (BT), a mixed mu/kappa-agonist, and nociceptin/orphanin FQ (N/OFQ), which binds to an ORL1 receptor. BT and N/OFQ completely blocked acquisition of CTA. MOR alleviated but did not eliminate the aversive effects. Each of the opioids decreased LiCl-induced activation of NTS neurons as well as OT and VP cells in the PVN and SON. We conclude that opioids antagonize aversive properties of LiCl, presumably by suppressing activation of pathways that encompass OT and VP cells and NTS neurons.


Subject(s)
Analgesics, Opioid/pharmacology , Avoidance Learning/drug effects , Lithium Chloride/pharmacology , Neurons/physiology , Oxytocin/physiology , Paraventricular Hypothalamic Nucleus/physiology , Supraoptic Nucleus/physiology , Taste , Vasopressins/physiology , Animals , Butorphanol/pharmacology , Male , Morphine/pharmacology , Neurons/drug effects , Opioid Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL
...