Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Eur Neuropsychopharmacol ; 29(11): 1235-1249, 2019 11.
Article in English | MEDLINE | ID: mdl-31519469

ABSTRACT

Amphetamine (AMPH), an appetite suppressant, alters expression levels of neuropeptide Y (NPY) and cocaine- and amphetamine-regulated transcript (CART) in the hypothalamus. This study explored the potential role of cJun-N-terminal kinases (JNK) in appetite control, mediated by reactive oxygen species (ROS) and activator protein-1 (AP-1) in AMPH-treated rats. Rats were given AMPH daily for 4 days. Changes in feeding behavior and expression levels of hypothalamic NPY, CART, cFos, cJun, phosphorylated JNK (pJNK), as well as those of anti-oxidative enzymes, including superoxide dismutase (SOD), glutathione peroxidase (GP) and glutathione S-transferase (GST), were examined and compared. Following AMPH treatment, food intake and NPY expression decreased, whereas the other proteins expression and AP-1/DNA binding activity increased. Both cerebral cJun inhibition and ROS inhibition attenuated AMPH anorexia and modified detected protein, revealing a crucial role for AP-1 and ROS in regulating AMPH-induced appetite control. Moreover, both pJNK/CART and SOD/CART activities detected by double immunofluorescent staining increased in hypothalamic arcuate nucleus in AMPH-treated rats. The results suggested that pJNK/AP-1 signaling and endogenous anti-oxidants participated in regulating NPY/CART-mediated appetite control in rats treated with AMPH. These findings advance understanding of the molecular mechanism underlying the role of pJNK/AP-1 and oxidative stress in NPY/CART-mediated appetite suppression in AMPH-treated rats.


Subject(s)
Appetite Regulation/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Neuropeptide Y/physiology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/physiology , Amphetamine/pharmacology , Animals , Anthracenes/administration & dosage , Anthracenes/pharmacology , Antioxidants/metabolism , Appetite Regulation/drug effects , Feeding Behavior/drug effects , Fluorescent Antibody Technique , Hypothalamus/metabolism , Hypothalamus/physiology , Infusions, Intraventricular , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Nerve Tissue Proteins/metabolism , Neuropeptide Y/biosynthesis , Rats , Signal Transduction/physiology , Transcription Factor AP-1/metabolism
2.
Physiol Behav ; 212: 112654, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31430441

ABSTRACT

Ketogenic diets (KDs) are high-fat, low-carbohydrate diets that have been used therapeutically for decades, most notably for the treatment of epilepsy and diabetes. Recent data, however, suggest that KD may impart protective effects on mood disorders. The current experiments test the hypothesis that KDs can protect from stress-induced symptoms of mood disorders. To test this, we assessed behavioral and neuroendocrine effects of KD in male and female Long Evans rats. Animals experienced three weeks of chronic mild stress (CMS) while consuming KD or control chow (CH). Body weight and food intake data were recorded daily and behaviors were assayed after three weeks. Plasma beta-hydroxybutyrate (ßHB), corticosterone (CORT) and interleukin-1 beta (IL-1ß) were measured after behavioral testing, along with hypothalamic corticotropin-releasing hormone (CRH) and neuropeptide Y (NPY) mRNA expression. CMS induced weight loss in the CH groups, however the KD-fed rats were resistant to CMS-induced weight loss. Female rats fed KD were protected from CMS-induced reductions in plasma CORT and hypothalamic NPY expression. Collectively, these data suggest protective potential of KDs against chronic stress, particularly in females.


Subject(s)
Diet, Ketogenic , Stress, Psychological/physiopathology , Weight Loss/physiology , 3-Hydroxybutyric Acid/blood , Animals , Behavior, Animal/physiology , Body Weight , Corticosterone/blood , Corticotropin-Releasing Hormone/biosynthesis , Eating , Female , Hypothalamus/metabolism , Interleukin-1beta/blood , Male , Neuropeptide Y/biosynthesis , Rats , Rats, Long-Evans , Sex Characteristics , Stress, Psychological/metabolism
3.
Eur Neuropsychopharmacol ; 29(4): 482-492, 2019 04.
Article in English | MEDLINE | ID: mdl-30878321

ABSTRACT

PTSD is heterogeneous disorder that can be long lasting and often has delayed onset following exposure to a traumatic event. Therefore, it is important to take a staging approach to evaluate progression of biological mechanisms of the disease. Here, we begin to evaluate the temporal trajectory of changes following exposure to traumatic stressors in the SPS rat PTSD model. The percent of animals displaying severe anxiety on EPM increased from 17.5% at one week to 57.1% two weeks after SPS stressors, indicating delayed onset or progressive worsening of the symptoms. The LC displayed prolonged activation, and dysbalance of the CRH/NPY systems, with enhanced CRHR1 gene expression, coupled with reduced mRNAs for NPY and Y2R. In the mediobasal hypothalamus, increased CRH mRNA levels were sustained, but there was a flip in alterations of HPA regulatory molecules, GR and FKBP5 and Y5 receptor at two weeks compared to one week. Two weeks after SPS, intranasal NPY at 300 µg/rat, but not 150 µg which was effective after one week, reversed SPS triggered elevated anxiety. It also reversed SPS elicited depressive/despair symptoms and hyperarousal. Overall, the results reveal time-dependent progression in development of anxiety symptoms and molecular impairments in gene expression for CRH and NPY systems in LC and mediobasal hypothalamus by SPS. With longer time afterwards only a higher dose of NPY was effective in reversing behavioral impairments triggered by SPS, indicating that therapeutic approaches should be adjusted according to the degree of biological progression of the disorder.


Subject(s)
Gene Expression , Hypothalamus/metabolism , Locus Coeruleus/metabolism , Neuropeptide Y/pharmacology , Stress Disorders, Post-Traumatic/metabolism , Animals , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/biosynthesis , Male , Neuropeptide Y/biosynthesis , Rats , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Neuropeptide Y/biosynthesis , Tacrolimus Binding Proteins/biosynthesis , Time Factors
4.
J Comp Neurol ; 527(9): 1508-1526, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30666646

ABSTRACT

Orexin A (OXA) and neuropeptide Y (NPY) are two hypothalamic neuropeptides involved in the regulation of feeding behavior and food intake in all vertebrates. Accumulating evidences document that they undergo age-related modifications, with consequences on metabolism, sleep/wake disorders and progression of neurodegenerations. The present study addressed the age related changes in expression and distribution of orexin A (its precursor is also known as hypocretin-HCRT) and NPY, and their regulation by food intake in the short-lived vertebrate model Nothobranchius furzeri. Our experiments, conducted on male specimens, show that: (a) HCRT and OXA and NPY mRNA and protein are localized in neurons of diencephalon and optic tectum, as well as in numerous fibers projecting through the entire neuroaxis, and are colocalized in specific nuclei; (b) in course of aging, HCRT and NPY expressing neurons are localized also in telencephalon and rhombencephalon; (c) HCRT expressing neurons increased slightly in the diencephalic area of old animals and in fasted animals, whereas NPY increased sharply; (d) central HCRT levels are not regulated neither in course of aging nor by food intake; and (e) central NPY levels are augmented in course of aging, and regulated by food intake only in young. These findings represent a great novelty in the study of central orexinergic and NPY-ergic systems in vertebrates', demonstrating an uncommon and unprecedented described regulation of these two orexigenic neuropeptides.


Subject(s)
Aging/metabolism , Diencephalon/metabolism , Eating/physiology , Fundulidae/metabolism , Gene Expression Regulation/physiology , Hypothalamus/metabolism , Neuropeptide Y/biosynthesis , Orexins/biosynthesis , Amino Acid Sequence , Animals , Conserved Sequence , Fasting/metabolism , Fundulidae/genetics , In Situ Hybridization , Male , Neurons/metabolism , Neuropeptide Y/genetics , Orexins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Superior Colliculi/metabolism
5.
J Neuroendocrinol ; 29(1)2017 01.
Article in English | MEDLINE | ID: mdl-27874965

ABSTRACT

The Siberian hamster (Phodopus sungorus) is a seasonal mammal, exhibiting a suite of physiologically and behaviourally distinct traits dependent on the time of year and governed by changes in perceived day length (photoperiod). These attributes include significant weight loss, reduced food intake, gonadal atrophy and pelage change with short-day photoperiod as in winter. The central mechanisms driving seasonal phenotype change during winter are mediated by a reduced availability of hypothalamic triiodothyronine (T3), although the downstream mechanisms responsible for physiological and behavioural changes are yet to be fully clarified. With access to a running wheel (RW) in short photoperiod, Siberian hamsters that have undergone photoperiod-mediated weight loss over-ride photoperiod-drive for reduced body weight and regain weight similar to a hamster held in long days. These changes occur despite retaining the majority of hypothalamic gene expression profiles appropriate for short-day hamsters. Utilising the somatostatin agonist pasireotide, we recently provided evidence for an involvement of the growth hormone (GH) axis in the seasonal regulation of bodyweight. In the present study, we employed pasireotide to test for the possible involvement of the GH axis in RW-induced body weight regulation. Pasireotide successfully inhibited exercise-stimulated growth in short-day hamsters and this was accompanied by altered hypothalamic gene expression of key GH axis components. Our data provide support for an involvement of the GH axis in the RW response in Siberian hamsters.


Subject(s)
Body Weight/drug effects , Motor Activity/drug effects , Receptors, Somatotropin/biosynthesis , Somatostatin/analogs & derivatives , Animals , Body Composition/drug effects , Cricetinae , Eating , Growth Hormone-Releasing Hormone/biosynthesis , Hypothalamus/metabolism , Iodide Peroxidase/biosynthesis , Male , Neuropeptide Y/biosynthesis , Organ Size/drug effects , Phodopus , Photoperiod , Pro-Opiomelanocortin/biosynthesis , Somatostatin/agonists , Somatostatin/biosynthesis , Somatostatin/pharmacology
6.
Planta Med ; 82(1-2): 84-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26756819

ABSTRACT

Fragrance in the air and odours of foods and drinks are reported to affect feeding behaviours of humans and other animals. Many previous studies focusing on the relationship between fragrance and appetite have described a reduction of food intake by fragrance administration to help prevent lifestyle diseases. Aromatic herbal medicines, such as cinnamon bark and fennel fruit, are considered to have appetite-enhancing effects and they are often blended in stomachics for relief of asitia and gastric distress in Japan. These fragrant herbal medicines contain many essential oils and their fragrances are hypothesised to be active substances. In this study, food intake and the expression of neuropeptide Y and proopiomelanocortin in the hypothalamus after inhalation of fragrant compounds or essential oils were investigated in mice. Food intake was increased 1.2-fold and the neuropeptide Y mRNA expression in the hypothalamus was increased significantly in mice that inhaled trans-cinnamaldehyde, benzylacetone or 1-phenyl-2-butanone, compared with the control group. These compounds might be effective for treating loss of appetite (anorexia) or eating disorders in elderly and infirm people via a non-invasive route of administration, namely, inhalation.


Subject(s)
Acetone/analogs & derivatives , Acrolein/analogs & derivatives , Butanones/pharmacology , Eating/drug effects , Hypothalamus/drug effects , Acetone/pharmacology , Acrolein/pharmacology , Administration, Inhalation , Animals , Hypothalamus/metabolism , Male , Mice , Neuropeptide Y/biosynthesis , Odorants , Oils, Volatile , Pro-Opiomelanocortin/biosynthesis
7.
Physiol Behav ; 153: 47-55, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26485293

ABSTRACT

We aimed to evaluate the effects of maternal and/or paternal obesity on offspring body mass, leptin signaling, appetite-regulating neurotransmitters and local inflammatory markers. C57BL/6 mice received standard chow (SC, lean groups) or high-fat diet (HF, obese groups) starting from one month of age. At three months, HF mice became obese relative to SC mice. They were then mated as follows: lean mother and lean father, lean mother and obese father, obese mother and lean father, and obese mother and obese father. The offspring received the SC diet from weaning until three months of age, when they were sacrificed. In the offspring, paternal obesity did not lead to changes in the Janus kinase (JAK)/signal transducer and activation of the transcription (STAT) pathway or feeding behavior but did induce hypothalamic inflammation. On the other hand, maternal obesity resulted in increased weight gain, hyperleptinemia, decreased leptin OBRb receptor expression, JAK/STAT pathway impairment, and increased SOCS3 signaling in the offspring. In addition, maternal obesity elevated inflammatory markers and altered NPY and POMC expression in the hypothalamus. Interestingly, combined parental obesity exacerbated the deleterious outcomes compared to single-parent obesity. In conclusion, while maternal obesity is known to program metabolic changes and obesity in offspring, the current study demonstrated that obese fathers induce hypothalamus inflammation in offspring, which may contribute to the development of metabolic syndromes in adulthood.


Subject(s)
Hyperphagia/metabolism , Hypothalamus/metabolism , Inflammation Mediators/metabolism , Leptin/metabolism , Obesity/metabolism , Parents , Signal Transduction , Animals , Body Weight , Diet, High-Fat , Energy Intake , Fathers , Female , Janus Kinase 1/metabolism , Leptin/blood , Male , Mice , Mothers , Neuropeptide Y/biosynthesis , Obesity/chemically induced , Obesity/physiopathology , Pro-Opiomelanocortin/biosynthesis , Receptors, Leptin/biosynthesis , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
8.
J Neuroendocrinol ; 27(12): 872-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26442444

ABSTRACT

Ghrelin, a gut hormone originating from the post-translational cleavage of preproghrelin, is the endogenous ligand of growth hormone secretagogue receptor 1a (GHS-R1a). Within the growth hormone (GH) axis, the biological activity of ghrelin requires octanoylation by ghrelin-O-acyltransferase (GOAT), conferring selective binding to the GHS-R1a receptor via acylated ghrelin. Complete loss of preproghrelin-derived signalling (through deletion of the Ghrl gene) contributes to a decline in peak GH release; however, the selective contribution of endogenous acyl-ghrelin to pulsatile GH release remains to be established. We assessed the pulsatile release of GH in ad lib. fed male germline goat(-/-) mice, extending measures to include mRNA for key hypothalamic regulators of GH release, and peripheral factors that are modulated relative to GH release. The amount of GH released was reduced in young goat(-/-) mice compared to age-matched wild-type mice, whereas pulse frequency and irregularity increased. Altered GH release did not coincide with alterations in hypothalamic Ghrh, Srif, Npy or Ghsr mRNA expression, or pituitary GH content, suggesting that loss of Goat does not compromise canonical mechanisms that contribute to pituitary GH production and release. Although loss of Goat resulted in an irregular pattern of GH release (characterised by an increase in the number of GH pulses observed during extended secretory events), this did not contribute to a change in the expression of sexually dimorphic GH-dependent liver genes. Of interest, circulating levels of insulin-like growth factor (IGF)-1 were elevated in goat(-/-) mice. This rise in circulating levels of IGF-1 was correlated with an increase in GH pulse frequency, suggesting that sustained or increased IGF-1 release in goat(-/-) mice may occur in response to altered GH release patterning. Our observations demonstrate that germline loss of Goat alters GH release and patterning. Although the biological relevance of altered GH secretory patterning remains unclear, we propose that this may contribute to sustained IGF-1 release and growth in goat(-/-) mice.


Subject(s)
Acyltransferases/deficiency , Acyltransferases/physiology , Growth Hormone/metabolism , Acyltransferases/genetics , Animals , Growth Hormone-Releasing Hormone/biosynthesis , Hypothalamus/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Membrane Proteins , Mice , Mice, Knockout , Neuropeptide Y/biosynthesis , Receptors, Ghrelin/biosynthesis , Somatostatin/biosynthesis
9.
J Mol Endocrinol ; 55(2): 95-106, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26162607

ABSTRACT

Neuromedin U (NMU) and neuromedin S (NMS) play inhibitory roles in the regulation of food intake and energy homeostasis in mammals. However, their functions are not clearly established in teleost fish. In the present study, nmu and nms homologs were identified in several fish species. Subsequently, their cDNA sequences were cloned from the orange-spotted grouper (Epinephelus coioides). Sequence analysis showed that the orange-spotted grouper Nmu proprotein contains a 21-amino acid mature Nmu peptide (Nmu-21). The Nms proprotein lost the typical mature Nms peptide, but it retains a putative 34-amino acid peptide (Nmsrp). In situ hybridization revealed that nmu- and nms-expressing cells are mainly localized in the hypothalamic regions associated with appetite regulation. Food deprivation decreased the hypothalamic nmu mRNA levels but induced an increase of nms mRNA levels. Periprandial expression analysis showed that hypothalamic expression of nmu increased significantly at 3 h post-feeding, while nms expression was elevated at the normal feeding time. I.p. injection of synthetic Nmu-21 peptide suppressed the hypothalamic neuropeptide y (npy) expression, while Nmsrp administration significantly increased the expression of npy and orexin in orange-spotted grouper. Furthermore, the mRNA levels of LH beta subunit (lhß) and gh in the pituitary were significantly down-regulated after Nmu-21 peptide administration, while Nmsrp was able to significantly stimulate the expression of FSH beta subunit (fshß), prolactin (prl), and somatolaction (sl). Our results indicate that nmu and nms possess distinct neuroendocrine functions and pituitary functions in the orange spotted grouper.


Subject(s)
Bass/genetics , Energy Metabolism/genetics , Fish Proteins/genetics , Neuropeptide Y/genetics , Neuropeptides/genetics , Amino Acid Sequence , Animals , Appetite/genetics , Base Sequence , Cloning, Molecular , Eating/genetics , Follicle Stimulating Hormone, beta Subunit/biosynthesis , Hypothalamus/cytology , Hypothalamus/metabolism , In Situ Hybridization , Luteinizing Hormone, beta Subunit/genetics , Molecular Sequence Data , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Orexins/biosynthesis , Pituitary Gland/metabolism , Prolactin/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sequence Analysis, DNA , Starvation/genetics
10.
Biosci Biotechnol Biochem ; 79(8): 1342-9, 2015.
Article in English | MEDLINE | ID: mdl-25952775

ABSTRACT

We previously found that daidzein decreased food intake in female rats. The present study aimed to elucidate the relationship between dynamics of appetite-mediated neuropeptides and the anorectic effect of daidzein. We examined appetite-mediated gene expression in the hypothalamus and small intestine during the 3 meals per day feeding method. Daidzein had an anorectic effect specifically at the second feeding. Neuropeptide-Y (NPY) and galanin mRNA levels in the hypothalamus were significantly higher after feeding in the control but not in the daidzein group, suggesting that daidzein attenuated the postprandial increase in NPY and galanin expression. The daidzein group had higher corticotrophin-releasing hormone (CRH) mRNA levels in the hypothalamus after feeding, and increased cholelcystokinin (CCK) mRNA levels in the small intestine, suggesting that CCK is involved in the hypothalamic regulation of this anorectic effect. Therefore, daidzein may induce anorexia by suppressing expression of NPY and galanin and increasing expression of CRH in the hypothalamus.


Subject(s)
Anorexia/genetics , Appetite/genetics , Eating/genetics , Galanin/biosynthesis , Neuropeptide Y/biosynthesis , Animals , Anorexia/pathology , Appetite/physiology , Body Weight , Eating/drug effects , Feeding Methods , Female , Galanin/genetics , Gene Expression Regulation/drug effects , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Isoflavones/administration & dosage , Neuropeptide Y/genetics , RNA, Messenger/biosynthesis , Rats , Receptors, Cholecystokinin/biosynthesis , Receptors, Corticotropin-Releasing Hormone/biosynthesis
11.
Am J Physiol Endocrinol Metab ; 306(11): E1284-91, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24735891

ABSTRACT

CCK and leptin are anorectic hormones produced in the small intestine and white adipose tissue, respectively. Investigating how these hormones act together as an integrated anorectic signal is important for elucidating the mechanisms by which energy balance is maintained. We found here that coadministration of subthreshold CCK and leptin, which individually have no effect on feeding, dramatically reduced food intake in rats. Phosphorylation of AMP-activated protein kinase (AMPK) in the hypothalamus significantly decreased after coinjection of CCK and leptin. In addition, coadministration of these hormones significantly increased mRNA levels of anorectic cocaine- and amphetamine-regulated transcript (CART) and thyrotropin-releasing hormone (TRH) in the hypothalamus. The interactive effect of CCK and leptin on food intake was abolished by intracerebroventricular preadministration of the AMPK activator AICAR or anti-CART/anti-TRH antibodies. These findings indicate that coinjection of CCK and leptin reduces food intake via reduced AMPK phosphorylation and increased CART/TRH in the hypothalamus. Furthermore, by using midbrain-transected rats, we investigated the role of the neural pathway from the hindbrain to the hypothalamus in the interaction of CCK and leptin to reduce food intake. Food intake reduction induced by coinjection of CCK and leptin was blocked in midbrain-transected rats. Therefore, the neural pathway from hindbrain to hypothalamus plays an important role in transmitting the anorectic signals provided by coinjection of CCK and leptin. Our findings give further insight into the mechanisms of feeding and energy balance.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Cholecystokinin/pharmacology , Eating/drug effects , Hypothalamus/metabolism , Leptin/pharmacology , Nerve Tissue Proteins/biosynthesis , Thyrotropin-Releasing Hormone/biosynthesis , Agouti-Related Protein/biosynthesis , Animals , Blotting, Western , Hypothalamus/drug effects , Hypothalamus/enzymology , Injections, Intraventricular , Male , Mesencephalon/physiology , Neuropeptide Y/biosynthesis , Phosphorylation , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/biosynthesis
12.
J Biol Chem ; 289(7): 4055-69, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24366864

ABSTRACT

CTRP4 is a unique member of the C1q family, possessing two tandem globular C1q domains. Its physiological function is poorly defined. Here, we show that CTRP4 is an evolutionarily conserved, ∼34-kDa secretory protein expressed in the brain. In human, mouse, and zebrafish brain, CTRP4 expression begins early in development and is widespread in the central nervous system. Neurons, but not astrocytes, express and secrete CTRP4, and secreted proteins form higher-order oligomeric complexes. CTRP4 is also produced by peripheral tissues and circulates in blood. Its serum levels are increased in leptin-deficient obese (ob/ob) mice. Functional studies suggest that CTRP4 acts centrally to modulate energy metabolism. Refeeding following an overnight fast induced the expression of CTRP4 in the hypothalamus. Central administration of recombinant protein suppressed food intake and altered the whole-body energy balance in both chow-fed and high-fat diet-fed mice. Suppression of food intake by CTRP4 is correlated with a decreased expression of orexigenic neuropeptide (Npy and Agrp) genes in the hypothalamus. These results establish CTRP4 as a novel nutrient-responsive central regulator of food intake and energy balance.


Subject(s)
Adipokines/metabolism , Body Weight/physiology , Cytokines/metabolism , Eating/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Zebrafish/metabolism , Adipokines/genetics , Agouti-Related Protein/biosynthesis , Agouti-Related Protein/genetics , Animals , Cytokines/genetics , Humans , Male , Mice , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , Protein Structure, Tertiary , Rats , Zebrafish/genetics
13.
J Neurochem ; 129(2): 297-303, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24266392

ABSTRACT

Hypothalamic appetite regulators neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) are modulated by glucose. This study investigated how maternal obesity disturbs glucose regulation of NPY and POMC, and whether this deregulation is linked to abnormal hypothalamic glucose uptake-lactate conversion. As post-natal high-fat diet (HFD) can exaggerate the effects of maternal obesity, its additional impact was also investigated. Female Sprague Dawley rats were fed a HFD (20 kJ/g) to model maternal obesity. At weaning, male pups were fed chow or HFD. At 9 weeks, in vivo hypothalamic NPY and POMC mRNA responses to acute hyperglycemia were measured; while hypothalami were glucose challenged in vitro to assess glucose uptake-lactate release and related gene expression. Maternal obesity dampened in vivo hypothalamic NPY response to acute hyperglycemia, and lowered in vitro hypothalamic glucose uptake and lactate release. When challenged with 20 mM glucose, hypothalamic glucose transporter 1, monocarboxylate transporters, lactate dehydrogenase-b, NPY and POMC mRNA expression were down-regulated in offspring exposed to maternal obesity. Post-natal HFD consumption reduced in vitro lactate release and monocarboxylate transporter 2 mRNA, but increased POMC mRNA levels when challenged with 20 mM glucose. Overall, maternal obesity produced stronger effects than post-natal HFD consumption to impair hypothalamic glucose metabolism. However, they both disturbed NPY response to hyperglycemia, potentially leading to hyperphagia.


Subject(s)
Brain Chemistry/physiology , Glucose/metabolism , Hyperglycemia/physiopathology , Obesity/metabolism , Prenatal Exposure Delayed Effects/metabolism , Adiposity/physiology , Animals , Cohort Studies , Female , Glucose/pharmacology , Glucose Transporter Type 1/metabolism , Growth/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Neuropeptide Y/biosynthesis , Pregnancy , Pro-Opiomelanocortin/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , Triglycerides/blood
14.
Tissue Cell ; 45(6): 452-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24138942

ABSTRACT

Neuropeptide Y (NPY) and orexin are neuropeptides involved in the regulation of feeding in vertebrates. In this study we determined the NPY and orexin mRNA tissue expression and their immunoreactivity distribution in both preoptic area and hypothalamus, regions involved in the regulation of feeding behavior. Both peptides presented a wide expression in all tissues examined. The NPY-immunoreactive (ir) cells were localized in the ventral nucleus posterioris periventricularis (NPPv) and numerous ir-NPY fibers were found in the nucleus lateralis tuberis (NLT), the nucleus recess lateralis (NRL) and the neurohypophysis. Ir-orexin cells were observed in the NPPv, dorsal NLT, ventral NLT, lateral NLT (NLTl) and the lateral NRL. Ir-orexin fibers were widespread distributed along all the hypothalamus, especially in the NLTl. Additionally, we observed the presence of ir-orexin immunostaining in adenohypophyseal cells, especially in somatotroph cells and the presence of a few ir-orexin-A fibers in the neurohypophysis. In conclusion, both peptides have an ubiquitous mRNA tissue expression and are similarly distributed in the hypothalamus and preoptic area of Cichlasoma dimerus. The presence of ir-orexin in adenohypohyseal cells and the presence of ir-orexin and NPY fibers in the neurohypophysis suggest that both peptides may play an important neuroendocrine role in anterior pituitary.


Subject(s)
Cichlids/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Animals , Cichlids/genetics , Orexins , Preoptic Area/metabolism
15.
Endocrinology ; 154(10): 3589-98, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23861376

ABSTRACT

Fifteen years ago orexins were identified as central regulators of energy homeostasis. Since then, that concept has evolved considerably and orexins are currently considered, besides orexigenic neuropeptides, key modulators of sleep-wake cycle and neuroendocrine function. Little is known, however, about the effect of the neuroendocrine milieu on orexins' effects on energy balance. We therefore investigated whether hypothalamic-pituitary axes have a role in the central orexigenic action of orexin A (OX-A) by centrally injecting hypophysectomized, adrenalectomized, gonadectomized (male and female), hypothyroid, and GH-deficient dwarf rats with OX-A. Our data showed that the orexigenic effect of OX-A is fully maintained in adrenalectomized and gonadectomized (females and males) rats, slightly reduced in hypothyroid rats, and totally abolished in hypophysectomized and dwarf rats when compared with their respective vehicle-treated controls. Of note, loss of the OX-A effect on feeding was associated with a blunted OX-A-induced increase in the expression of either neuropeptide Y or its putative regulator, the transcription factor cAMP response-element binding protein, as well as its phosphorylated form, in the arcuate nucleus of the hypothalamus of hypophysectomized and dwarf rats. Overall, this evidence suggests that the orexigenic action of OX-A depends on an intact GH axis and that this neuroendocrine feedback loop may be of interest in the understanding of orexins action on energy balance and GH deficiency.


Subject(s)
Appetite Regulation , Growth Hormone/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Pituitary Gland/metabolism , Receptors, Somatotropin/metabolism , Adrenalectomy/adverse effects , Animals , Castration/adverse effects , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP Response Element-Binding Protein/metabolism , Dwarfism, Pituitary/metabolism , Dwarfism, Pituitary/physiopathology , Feeding Behavior , Female , Hypophysectomy/adverse effects , Hypothalamus/metabolism , Hypothyroidism/metabolism , Hypothyroidism/physiopathology , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins/administration & dosage , Male , Neuropeptide Y/biosynthesis , Neuropeptide Y/metabolism , Neuropeptides/administration & dosage , Orexins , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley
16.
Peptides ; 46: 102-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23756158

ABSTRACT

Neuropeptide Y (NPY) is a potent orexigenic neuropeptide implicated in appetite regulation in mammals. However, except for teleost fish such as the goldfish and zebrafish, the involvement of NPY in the regulation of feeding in non-mammalian vertebrates has not been well studied. Anuran amphibian larvae feed and grow during the pre- and pro-metamorphic stages, but, thereafter they stop feeding as the metamorphic climax approaches. Therefore, orexigenic factors seem to play important roles in pre- and pro-metamorphic larvae. We investigated the role of NPY in food intake using bullfrog larvae including pre- and pro-metamorphic stages, and examined the effect of feeding status on the expression level of the NPY transcript in the hypothalamus. NPY mRNA levels in hypothalamus specimens obtained from larvae that had been fasted for 3 days were higher than those in larvae that had been fed normally. We then investigated the effect of intracerebroventricular (ICV) administration of NPY on food intake in the larvae. Cumulative food intake was significantly increased by ICV administration of NPY (5 and 10 pmol/g body weight, BW) during a 15-min observation period. The NPY-induced orexigenic action (10 pmol/g BW) was blocked by treatment with a NPY Y1 receptor antagonist, BIBP-3226 (100 pmol/g BW). These results indicate that NPY acts as an orexigenic factor in bullfrog larvae.


Subject(s)
Appetite Regulation/physiology , Eating , Feeding Behavior/physiology , Neuropeptide Y/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Arginine/analogs & derivatives , Arginine/pharmacology , Fasting , Hypothalamus/metabolism , Larva/metabolism , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , RNA, Messenger/biosynthesis , Rana catesbeiana , Receptors, Neuropeptide Y/antagonists & inhibitors
17.
Neuropeptides ; 47(3): 139-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23490004

ABSTRACT

The hypothalamic neurochemicals neuropeptide Y (NPY), orexin-A (ORX), and oxytocin (OXY) exert glucoregulatory effects upon intracerebral administration, findings that support their potential function within neural pathways that maintain glucostasis. Current understanding of how these neurotransmitter systems respond to the diabetes mellitus complication, insulin-induced hypoglycemia, is limited to knowledge of neuropeptide gene transcriptional reactivity. We investigated the hypothesis that hypoglycemia elicits hypothalamic site-specific alterations in levels of these neurochemicals, and that adjustments in local neurotransmitter availability may be regulated by catecholaminergic (CA) input from the caudal dorsomedial hindbrain. The arcuate (ARH) and paraventricular (PVH) hypothalamic nuclei and lateral hypothalamic area (LHA) were each microdissected from adult male rats pretreated by caudal fourth ventricular administration of the selective CA neurotoxin, 6-hydroxydopamine (6-OHDA), or vehicle prior to insulin (INS)-induced hypoglycemia. Hypoglycemia stimulated ARH NPY gene expression and NPY accumulation in the ARH and LHA, but not PVH. 6-OHDA pretreatment did not modify the positive NPY mRNA response to INS, but blunted hypoglycemic augmentation of ARH and LHA NPY content while increasing PVH NPY levels in response to hypoglycemia. INS-treated rats exhibited diminished LHA ORX gene expression and increased [ARH; LHA] or decreased [PVH] tissue ORX protein levels. 6-OHDA+INS animals showed a comparable decline in ORX transcripts, but attenuated augmentation of ARH and LHA ORX content and elevated PVH ORX levels. OT mRNA and protein were respectively decreased or unchanged during hypoglycemia, responses that were uninfluenced by hindbrain CA nerve cell destruction. These results illustrate divergent adjustments in glucoregulatory neurotransmitter gene expression and site-specific protein accumulation in the hypothalamus during hypoglycemia. Evidence that 6-OHDA pretreatment does not modify NPY or ORX transcriptional reactivity to hypoglycemia, but alters hypoglycemic patterns of NPY and ORX accretion implicates dorsomedial hindbrain CA neurons in regulation of translation/post-translational processing and site-specific availability of these neurotransmitters in the hypothalamus during hypoglycemia.


Subject(s)
Catecholamines/metabolism , Glucose/metabolism , Hypoglycemia/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Oxytocin/biosynthesis , Rhombencephalon/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Hydroxydopamines/pharmacology , Hypoglycemia/chemically induced , Hypoglycemic Agents , Immunohistochemistry , Insulin , Intracellular Signaling Peptides and Proteins/genetics , Male , Neuropeptide Y/genetics , Neuropeptides/genetics , Neurotoxins/metabolism , Neurotransmitter Agents/metabolism , Orexins , Oxytocin/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Sympathectomy, Chemical
18.
J Med Chem ; 56(5): 1811-29, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23384387

ABSTRACT

Newly synthesized acylethanolamide derivatives oleoyl-L-valinolamide (1), oleoyl-D-valinolamide (2), elaidoyl-L-valinolamide (3), elaidoyl-D-valinolamide (4) stearoyl-L-valinolamide (5), and palmitoyl-L-valinolamide (6) were investigated in mice as antiobesity compounds. Compounds 1, 2, 5, 6 significantly decreased body weight by 6.57% following eight injections of 1 mg/kg i.p. during 39 days, while 3 and 4 showed no such activity. Receptor binding indicated that no compound activated CB1, CB2, PPARα, or TRPV1 receptors. Hypothalamic RT-PCR showed that mRNA expression of the anorexigenic genes POMC and CART was up-regulated by 1, 2, 5 and 1, 2, respectively, while that of the orexigenic genes NPY and CaMKK2 was down-regulated by the respective compounds 1, 5, 6 and 1, 2, 5. Oleoyl-L-valinolamide enhances anorectic pathways and lead to decreased glucose levels, enhanced locomotor activity, and improved cognition. Effects of oleoyl-L-valinolamide on weight were dose-dependent, and it could be given orally. 1, 2, 4, 5 down-regulated FAAH mRNA expression.


Subject(s)
Amides/pharmacology , Anti-Obesity Agents/chemical synthesis , Body Weight/drug effects , Hypothalamus/metabolism , Neuropeptide Y/antagonists & inhibitors , Oleic Acids/chemical synthesis , Pro-Opiomelanocortin/biosynthesis , Valine/analogs & derivatives , Animals , Anti-Obesity Agents/pharmacology , Eating/drug effects , Ethanolamines/pharmacology , Female , Hypothalamus/drug effects , Mice , Mice, Inbred C57BL , Mice, Obese , Neuropeptide Y/biosynthesis , Nuclear Magnetic Resonance, Biomolecular , Oleic Acids/pharmacology , Valine/chemical synthesis , Valine/pharmacology
19.
J Chem Neuroanat ; 46(1-2): 45-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23085194

ABSTRACT

The role of exogenous ghrelin in the regulation of neuropeptide Y (NPY) neuronal system in the hypothalamus of intact lambs has not been yet determined. The aim of present study was to investigate the effects of intracerebroventricular infusion of ghrelin or short fasting on the secretory activity of the NPY neurons in the hypothalamus of prepubertal female sheep. Animals (n=30) were randomly divided into three groups, two groups were fed standard diet and one group was fasted for 72h. One group fed standard diet and fasted group were infused to the 3rd ventricle of the brain with vehicle, while the remaining group fed standard diet was infused with ghrelin (25µg/120µl/h) for 6h during three consecutive days. Immediately after the treatment, tissues were collected. Parts of the brains were fixed in situ for further immunohistochemical analysis, and remaining parts were frozen for RT-PCR analysis. Both, fasting and ghrelin infusion elicited the same kind of changes in the mRNA and intra-neuronal levels of the NPY hypothalamic neurons. Namely, the expression of NPY mRNA in the medial basal hypothalamus and immunoreactivity of NPY in the arcuate and periventricular nuclei increased in fasted and standard fed with ghrelin's infusion groups compared to standard fed sheep (P<0.05). These data demonstrate that ghrelin takes part in the mechanisms linking the nutritional status with an activity of the hypothalamic NPY at the level of the central nervous system by stimulating NPY secretion in sheep.


Subject(s)
Fasting/metabolism , Gene Expression Regulation , Ghrelin/administration & dosage , Hypothalamus/metabolism , Neurons/metabolism , Neuropeptide Y/biosynthesis , Age Factors , Animals , Female , Hypothalamus/chemistry , Hypothalamus/drug effects , Infusions, Intraventricular , Neurons/chemistry , Random Allocation , Sheep, Domestic , Time Factors , Treatment Outcome , Up-Regulation/physiology
20.
Gen Comp Endocrinol ; 179(2): 184-95, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22898434

ABSTRACT

In this study, we characterized the neuropeptide Y (NPY) mRNA in snakeskin gourami (Trichogaster pectoralis) (TpNPY). TpNPY displayed characteristics typical of previously reported NPYs, and it exhibited a high degree of homology with the NPY proteins of other vertebrates. A phylogenetic analysis demonstrated that TpNPY was closely related to the NPYs found in the acanthomorpha and salmoniformes fish species. TpNPY was found to be ubiquitously expressed in all brain regions when assessed by real-time RT-PCR and in situ hybridization. In addition, a graded expression level of TpNPY was observed in peripheral tissues; for example, a moderate level of TpNPY was found in the gills, liver, kidney, stomach, intestine, spleen and gonads, while a low level of TpNPY was found in the muscle. The change in expression of TpNPY with respect to daily feeding habits was investigated in distinct brain regions, including the telencephalon, mesencephalon, metencephalon, and diencephalon. Fluctuations in the expression level of TpNPY were observed for a 24h post-prandial period. Except for the telencephalon, a reduction in TpNPY expression was found after a meal, while a peak level of TpNPY was observed 1h before the scheduled breakfast. Furthermore, there was a positive correlation between TpNPY and TpMC4R in the telencephalon and diencephalon throughout the circadian feeding cycle, which suggests that there is a connection between the function of NPY and the melanocortin system for the regulation of daily feeding. Fish brains were incubated with an MC4R antagonist (i.e., HS024), and the expression of TpNPY and TpMC4R was measured. Interestingly, there was a significant relationship between the expression of TpNPY and TpMC4R under the effects of HS024, which demonstrates that there are interactions between MC4R and NPY, particularly in a hyperphagic state.


Subject(s)
Eating/physiology , Neuropeptide Y/physiology , Perciformes/metabolism , Receptor, Melanocortin, Type 4/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Circadian Rhythm , DNA, Complementary/chemistry , Molecular Sequence Data , Neuropeptide Y/biosynthesis , Peptides, Cyclic/pharmacology , Phylogeny , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Sequence Alignment , Telencephalon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL