Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Peptides ; 170: 171112, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37918484

ABSTRACT

Growth differentiation factor-15 (GDF15) is a stress-activated cytokine that regulates cell growth and inflammatory and stress responses. We previously reported the role and regulation of GDF15 in pituitary corticotrophs. Dexamethasone increases Gdf15 gene expression levels and production. GDF15 suppresses adrenocorticotropic hormone synthesis in pituitary corticotrophs and subsequently mediates the negative feedback effect of glucocorticoids. Here, we analyzed corticotropin-releasing factor (Crf) promoter activity in hypothalamic 4B cells transfected with promoter-driven luciferase reporter constructs. The effects of time and GDF15 concentration on Crf mRNA levels were analyzed using quantitative real-time polymerase chain reaction. Glial cell-derived neurotrophic factor family receptor α-like (GFRAL) protein is expressed in 4B cells. GDF15 increased Crf promoter activity and Crf mRNA levels in 4B cells. The protein kinase A and C pathways also contributed to the GDF15-induced increase in Crf gene expression. GDF15 stimulates GFRAL, subsequently increasing the phosphorylation of AKT, an extracellular signal-related kinase, and the cAMP response element-binding protein. Therefore, GDF15-dependent pathways may be involved in regulating Crf expression under stressful conditions in hypothalamic cells.


Subject(s)
Corticotropin-Releasing Hormone , Growth Differentiation Factor 15 , Hypothalamus , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Growth Differentiation Factors/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Promoter Regions, Genetic , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , RNA, Messenger/metabolism , Animals , Rats , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/pharmacology , Humans
2.
Front Endocrinol (Lausanne) ; 14: 1266081, 2023.
Article in English | MEDLINE | ID: mdl-37900150

ABSTRACT

The hypothalamic type 2 corticotropin releasing hormone receptor (CRH-R2) plays critical roles in homeostatic regulation, particularly in fine tuning stress recovery. During acute stress, the CRH-R2 ligands CRH and urocortins promote adaptive responses and feeding inhibition. However, in rodent models of chronic stress, over-exposure of hypothalamic CRH-R2 to its cognate agonists is associated with urocortin 2 (Ucn2) resistance; attenuated cAMP-response element binding protein (CREB) phosphorylation and increased food intake. The molecular mechanisms involved in these altered CRH-R2 signalling responses are not well described. In the present study, we used the adult mouse hypothalamus-derived cell line mHypoA-2/30 to investigate CRH-R2 signalling characteristics focusing on gene expression of molecules involved in feeding and circadian regulation given the role of clock genes in metabolic control. We identified functional CRH-R2 receptors expressed in mHypoA-2/30 cells that differentially regulate CREB and AMP-activated protein kinase (AMPK) phosphorylation and downstream expression of the appetite-regulatory genes proopiomelanocortin (Pomc) and neuropeptide Y (Npy) in accordance with an anorexigenic effect. We studied for the first time the effects of Ucn2 on clock genes in native and in a circadian bioluminescence reporter expressing mHypoA-2/30 cells, detecting enhancing effects of Ucn2 on mRNA levels and rhythm amplitude of the circadian regulator Aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), which could facilitate anorexic responses in the activity circadian phase. These data uncover novel aspects of CRH-R2 hypothalamic signalling that might be important in regulation of circadian feeding during stress responses.


Subject(s)
Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone , Mice , Animals , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Urocortins/genetics , Urocortins/metabolism , Gene Expression , Neurons/metabolism
3.
Neurochem Res ; 48(7): 2175-2186, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36853481

ABSTRACT

Activating transcription factor 6 (ATF6) is an endoplasmic reticulum (ER) stress-regulated transcription factor that induces expression of major molecular chaperones in the ER. We recently reported that ATF6ß, a subtype of ATF6, promoted survival of hippocampal neurons exposed to ER stress and excitotoxicity, at least in part by inducing expression of calreticulin, an ER molecular chaperone with high Ca2+-binding capacity. In the present study, we demonstrate that ATF6ß deficiency in mice also decreases calreticulin expression and increases expression of glucose-regulated protein 78, another ER molecular chaperone, in emotional brain regions such as the prefrontal cortex (PFC), hypothalamus, hippocampus, and amygdala. Comprehensive behavioral analyses revealed that Atf6b-/- mice exhibit anxiety-like behavior in the light/dark transition test and hyperactivity in the forced swim test. Consistent with these results, PFC and hypothalamic corticotropin-releasing hormone (CRH) expression was increased in Atf6b-/- mice, as was circulating corticosterone. Moreover, CRH receptor 1 antagonism alleviated anxiety-like behavior in Atf6b-/- mice. These findings suggest that ATF6ß deficiency produces anxiety-like behavior and hyperactivity via a CRH receptor 1-dependent mechanism. ATF6ß could play a role in psychiatric conditions in the emotional centers of the brain.


Subject(s)
Calreticulin , Receptors, Corticotropin-Releasing Hormone , Mice , Animals , Receptors, Corticotropin-Releasing Hormone/metabolism , Calreticulin/metabolism , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Anxiety/metabolism , Corticosterone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Psychological/complications , Stress, Psychological/metabolism , Activating Transcription Factor 6/metabolism
4.
Genes Brain Behav ; 20(8): e12775, 2021 11.
Article in English | MEDLINE | ID: mdl-34672092

ABSTRACT

The endocannabinoid system is an important regulator of the hormonal and behavioral stress responses, which critically involve corticotropin-releasing factor (CRF) and its receptors. While it has been shown that CRF and the cannabinoid type 1 (CB1) receptor are co-localized in several brain regions, the physiological relevance of this co-expression remains unclear. Using double in situ hybridization, we confirmed co-localization in the piriform cortex, the lateral hypothalamic area, the paraventricular nucleus, and the Barrington's nucleus, albeit at low levels. To study the behavioral and physiological implications of this co-expression, we generated a conditional knockout mouse line that selectively lacks the expression of CB1 receptors in CRF neurons. We found no effects on fear and anxiety-related behaviors under basal conditions nor after a traumatic experience. Additionally, plasma corticosterone levels were unaffected at baseline and after restraint stress. Only acoustic startle responses were significantly enhanced in male, but not female, knockout mice. Taken together, the consequences of depleting CB1 in CRF-positive neurons caused a confined hyperarousal phenotype in a sex-dependent manner. The current results suggest that the important interplay between the central endocannabinoid and CRF systems in regulating the organism's stress response is predominantly taking place at the level of CRF receptor-expressing neurons.


Subject(s)
Receptor, Cannabinoid, CB1/metabolism , Reflex, Startle/genetics , Acoustic Stimulation , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , Piriform Cortex/cytology , Piriform Cortex/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Sex
5.
Horm Behav ; 135: 105044, 2021 09.
Article in English | MEDLINE | ID: mdl-34507241

ABSTRACT

Corticotropin-releasing factor (CRF) signaling through CRF receptor 1 (CRFR1) regulates autonomic, endocrine, and behavioral responses to stress, as well as behavioral changes during the maternal period. Previous work in our lab reported higher levels of CRFR1 in female, compared to male, mice within the rostral anteroventral periventricular nucleus (AVPV/PeN), a brain region involved in maternal behaviors. In this study, we used CRFR1-GFP reporter mice to investigate whether the reproductive status (postpartum vs. nulliparous) of acutely stressed females affects levels of CRFR1 in the AVPV/PeN and other regions involved in maternal functions. Compared to nulliparous, postpartum day 14 females showed increased AVPV/PeN CRFR1-GFP immunoreactivity and an elevated number of restraint stress-activated AVPV/PeN CRFR1 cells as assessed by immunohistochemical co-localization of CRFR1-GFP and phosphorylated CREB (pCREB). The medial preoptic area (MPOA) and paraventricular hypothalamus (PVN) of postpartum mice showed modest decreases in CRFR1-GFP immunoreactivity, while increased CRFR1-GFP/pCREB co-expressing cells were found in the PVN following restraint stress relative to nulliparous mice. Tyrosine hydroxylase (TH) and CRFR1-GFP co-localization was also assessed in the AVPV/PeN and other regions and revealed a decrease in co-localized neurons in the AVPV/PeN and ventral tegmental area of postpartum mice. Corticosterone analysis of restrained mice revealed blunted peak, but elevated recovery, levels in postpartum compared to nulliparous mice. Finally, we investigated projection patterns of AVPV/PeN CRFR1 neurons using female CRFR1-Cre mice and revealed dense efferent projections to several preoptic, hypothalamic, and hindbrain regions known to control stress-associated and maternal functions. Together, these findings contribute to our understanding of the neurobiology that might underlie changes in stress-related functions during the postpartum period.


Subject(s)
Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone , Animals , Corticotropin-Releasing Hormone/metabolism , Female , Humans , Hypothalamus/metabolism , Male , Mice , Postpartum Period , Preoptic Area/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 321(4): R603-R613, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34405712

ABSTRACT

Stress in vertebrates is mediated by the hypothalamus-pituitary-adrenal (in mammals)/interrenal (in fish) (HPA/I) axis, which produces the corticotropin-releasing factor (CRF), adrenocorticotropic hormone (ACTH), and corticosteroids, respectively. Nesfatin-1, a novel anorexigenic peptide encoded in the precursor nucleobindin-2 (NUCB2), is increasingly acknowledged as a peptide that influences the stress axis in mammals. The primary aim of this study was to characterize the putative effects of nesfatin-1 on the fish HPI axis, using goldfish (Carassius auratus) as an animal model. Our results demonstrated that nucb2/nesfatin-1 transcript abundance was detected in the HPI tissues of goldfish, with most abundant expression in the pituitary. NUCB2/nesfatin-1-like immunoreactivity was found in the goldfish hypothalamus, pituitary, and interrenal cells of the head kidney. GPCR12, a putative receptor for nesfatin-1, was also detected in the pituitary and interrenal cells. NUCB2/nesfatin-1-like immunoreactivity was observed in ACTH-expressing pituitary corticotrophs. Acute netting and restraint stress upregulated nucb2/nesfatin-1 mRNA levels in the forebrain, hypothalamus, and pituitary, as well as crf and crf-r1 expression in the forebrain and hypothalamus. Intraperitoneal and intracerebroventricular administration of nesfatin-1 increased cortisol release and hypothalamic crf mRNA levels, respectively. Finally, we found that nesfatin-1 significantly stimulated ACTH secretion from dispersed pituitary cells in vitro. Collectively, our data provide the first evidence showing that nesfatin-1 is a stress responsive peptide, which modulates the stress axis hormones in fish.


Subject(s)
Fish Proteins/metabolism , Goldfish/metabolism , Hypothalamus/metabolism , Kidney/metabolism , Nucleobindins/metabolism , Pituitary Gland/metabolism , Animals , Cells, Cultured , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Female , Fish Proteins/genetics , Goldfish/genetics , Male , Nucleobindins/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, G-Protein-Coupled/metabolism , Restraint, Physical
7.
Medicine (Baltimore) ; 100(19): e25854, 2021 May 14.
Article in English | MEDLINE | ID: mdl-34106630

ABSTRACT

BACKGROUND: Insomnia is a common disease associated with different nervous system stress response and endocrine disorders. It has been reported previously that abdominal vibration and ring massage therapy can significantly improve the symptoms of insomnia patients, enhance the activity of neurons. In addition, functional MRI (resting state brain functional magnetic resonance imaging [Rs_fMRI]) of the resting state brain test has proved that the functional connection between hypothalamus and parahippocampal gyrus could be significantly enhanced after abdominal massage treatment. It has been confirmed that there is possible involvement of brain-gut interaction effect in the treatment of insomnia, but there is a lack of research to elucidate the possible mechanisms of brain-gut interaction in the treatment of insomnia. The purpose of this study is to investigate the relationship between the hypothalamus and intestinal interaction in the treatment of insomnia by abdominal massage. METHODS AND DESIGN: A single blind randomized controlled trial will be conducted. Sixty chronic insomnia volunteers and 30 healthy volunteers will be recruited for this study. Sixty insomnia volunteers will be randomly divided into a drug group and a massage group, and 30 healthy volunteers will be assigned to the healthy group. The manipulation of the treatment group will be mainly carried out through abdominal rubbing and vibration massage, once a day, 30 min/time, 5 days for a course of treatment, and a total of 4 intervention courses will be carried out. Patients in the drug group will be given orally spleen-invigorating bolus, twice a day, 1 pill in the morning and 1 pill in the evening. The course of treatment will be carried for 5 days, and a total of 4 courses of treatment will be administered.The massage group will be compared with the healthy group and the drug group by Pittsburgh Sleep Index scale (PSQI), Hyperarousal scale (HAS), Hamilton Depression scale (HAMD), Fatigue scale-14 (FS-14), and Wechsler Adult Memory scale (WAIS) scales using to observe the sleep quality. Rs-fMRI will be used to observe various BOLD signals in the brain and compare the values of Reho, fALFF, and FC. MRS technology will be used to observe the contents of GABA and 5-HT in the hypothalamus. Additionally, the contents of cortical hormone releasing hormone (CRH), adrenocorticotropic hormone (ACTH), COR, GABA, NE, PGE2, and 5-HT in the serum will be also detected. The serum of each group will be taken for 1H nuclear magnetic resonance (1HNMR) metabolomics study to analyze the various common metabolites, differential metabolites, potential metabolic biomarkers, and metabolic pathways among the 3 groups. Finally, in combination with the brain functional imaging and brain spectrum, the potential mechanism of abdominal vibration and ring massage will be discussed. DISCUSSION: The results of this study will be used to possibly elaborate the various mechanisms of brain and intestine interaction in the treatment of insomnia by employing abdomen ring rubbing.


Subject(s)
Intestines/physiology , Massage/methods , Pituitary Hormone-Releasing Hormones/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Sleep Initiation and Maintenance Disorders/physiopathology , Sleep Initiation and Maintenance Disorders/therapy , Chronic Disease , Drugs, Chinese Herbal/therapeutic use , Humans , Magnetic Resonance Spectroscopy , Single-Blind Method , Sleep/physiology , Vibration/therapeutic use
8.
J Ethnopharmacol ; 275: 114069, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33794334

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Stress is a state of feeling that inhibits one from responding properly in the face of a threat. Agarwood smoke has been used in traditional medicine as a sedative anti-anxious, and anti-restless therapy. Its scent emitted from heat induces people to enter a stable state; however, the underlying molecular effect is still unclear. AIM OF THE STUDY: This study analyzed novel biological events and gene expression signatures induced by agarwood incense smoke in mice. MATERIALS AND METHODS: Incense smoke was produced by heating at 150 °C for 30 min in a headspace autosampler oven. We treated mice with exposure to incense smoke from Kynam agarwood for 45 min/day for 7 consecutive days. After a 7-day inhalation period, the potent agarwood smoke affected-indicators in serum were measured, and the RNA profiles of the mouse brains were analyzed by microarray to elucidate the biological events induced by agarwood incense smoke. RESULTS: Chemical profile analysis showed that the major component in the incense smoke of Kynam was 2-(2-phenylethyl) chromone (26.82%). Incense smoke from Kynam induced mice to enter a stable state and increased the levels of serotonin in sera. The emotion-related pathways, including dopaminergic synapse, serotonergic synapse, GABAergic synapse, long-term depression and neuroactive ligand-receptor interaction, were significantly affected by incense smoke. Moreover, the expression of Crhr2 and Chrnd genes, involved with neuroactive ligand-receptor interaction pathway, was upregulated by incense smoke. CONCLUSIONS: By a newly-established incense smoke exposure system, we first identified that anti-anxious and anti-depressant effects of agarwood incense smoke were likely associated with the increase of serotonin levels and multiple neuroactive pathways in mice.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Hypnotics and Sedatives/pharmacology , Plant Extracts/pharmacology , Serotonin/metabolism , Smoke/analysis , Wood/chemistry , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/chemistry , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Brain/drug effects , Emotions/drug effects , Flavonoids/pharmacology , Flavonoids/therapeutic use , Gene Expression/drug effects , Hypnotics and Sedatives/chemistry , Hypnotics and Sedatives/therapeutic use , Male , Medicine, Traditional , Mice, Inbred BALB C , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
9.
Neurosci Lett ; 736: 135282, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32738351

ABSTRACT

Beta-melanocyte-stimulating hormone (ß-MSH), when centrally injected, induces anorexigenic effects in rodents and chickens but its mechanism remains unclear. Thus, the primary goal of this research was to elucidate the hypothalamic mechanism using chickens. Intracerebroventricular injection of 0.3, 1.0 and 3.0 nmol of ß-MSH decreased food intake for 540 min. Expression of hypothalamic mRNAs were affected by ß-MSH injection, including corticotrophin-releasing factor (CRF) and its receptor subtype 1 (CRFR1), mesotocin (MT) and its receptor (MTR), pro-opiomelanocortin, cocaine- and amphetamine-regulated transcript (CART), growth hormone secretagogue receptor (GHSR) and neuropeptide Y (NPY) receptor subtype 5 (NPYR5). Within the arcuate nucleus, expressions of NPY, agouti-related peptide, MT and MTR were increased by ß-MSH injection. ß-MSH-treated chicks had more CRF, CRFR1, CRF receptor subtype 2, GHSR, NPY receptor subtype 1 (NPYR1) and NPYR5 mRNA but lower levels of CART and ghrelin, in the paraventricular nucleus. Greater amounts of mRNA for MTR, GHSR, NPYR1 and NPYR5 and less CRF expression were observed in the ventromedial hypothalamus. In conclusion, central injection of ß-MSH potently reduced food intake and was associated with changes in mRNA expression of some anorexigenic factors in a hypothalamic nucleus-specific manner.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Eating/drug effects , Hypothalamus/drug effects , Oxytocin/analogs & derivatives , Receptors, Corticotropin-Releasing Hormone/metabolism , beta-MSH/pharmacology , Animals , Chickens , Hypothalamus/metabolism , Injections, Intraventricular , Oxytocin/metabolism
10.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R338-R350, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31850818

ABSTRACT

Exposure to different stressors in utero is linked to adult diseases such as obesity and hypertension. In this study, the impact of prenatal infection (PNI) on adult body weight and cardiovascular function was evaluated using a naturally occurring rodent pathogen, Mycoplasma pulmonis (MP). Pregnant Sprague-Dawley rats were infected with MP on gestationalday 14 and gave birth naturally. Adult PNI offspring weighed more than controls, but resting mean arterial pressure (MAP) was unchanged. Subcutaneous injection of angiotensin II (10 µg/kg) elicited a rise in MAP that was greater in both male and female PNI offspring compared with controls (P < 0.03). The accompanying reflex bradycardia was similar to the controls, suggesting that PNI induced baroreflex dysfunction. Subcutaneous nicotine administration, a potent cardiorespiratory stimulus, also elicited a transient rise in MAP that was generally greater in the PNI group, but the change in MAP from baseline was only significant in the PNI females compared with controls (P < 0.03). Elevated body weight and cardiovascular reactivity in the PNI offspring was associated with an increase in the ratio of hypothalamic corticotrophin-releasing hormone receptors type 1 to type 2 gene expression in both sexes compared with controls. These findings support previous studies demonstrating that PNI induces alterations in cardiovascular function and body weight. Yet, unlike previous studies utilizing other models of PNI (e.g., endotoxin), MP PNI did not induce resting hypertension. Thus, our study provides a foundation for future studies evaluating the cardiovascular risks of offspring exposed to microbial challenges in utero.


Subject(s)
Angiotensin II/administration & dosage , Arterial Pressure/drug effects , Baroreflex/drug effects , Cardiovascular Diseases/etiology , Mycoplasma Infections/complications , Mycoplasma pulmonis/pathogenicity , Prenatal Exposure Delayed Effects , Age Factors , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Female , Gestational Age , Hypothalamus/metabolism , Hypothalamus/physiopathology , Injections, Subcutaneous , Male , Mycoplasma Infections/microbiology , Pregnancy , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Weight Gain
11.
Int J Mol Med ; 45(1): 53-60, 2020 01.
Article in English | MEDLINE | ID: mdl-31746413

ABSTRACT

The gut­brain interaction is associated with impaired duodenal mucosal integrity and low­grade inflammation, which have been proven to be important pathological mechanisms of functional dyspepsia (FD). Sini San (SNS) is a classical Chinese medicine used to treat FD, but its underlying mechanisms are poorly understood. The aim of the present study was to evaluate the effects of SNS on duodenal mucosal barrier injury and low­grade inflammation with FD, and to assess its potential molecular mechanisms on the brain­gut axis. FD rats were established using the iodoacetamide and tail­squeezed methods. The expression of corticotropin­releasing factor (CRF), CRF receptor 1 (CRF­R1) and CRF­R2, were determined by western blot analysis and/or immunohistochemistry (IHC). In addition, mast cell (MC) migration was assessed by IHC with an anti­tryptase antibody, and histamine concentration was quantified using ELISA. The mRNA expression levels of tryptase and protease­activated receptor 2 (PAR­2) were quantified using reverse transcription­quantitative PCR, and the protein expression levels of zona occludens protein 1 (ZO­1), junctional adhesion molecule 1 (JAM­1), ß­catenin and E­cadherin were determined via western blot analysis. It was demonstrated that the expression level of CRF was downregulated in the central nervous system and duodenum following SNS treatment, and that SNS modulated the expression of both CRF­R1 and CRF­R2. In addition, SNS suppressed MC infiltration and the activity of the tryptase/PAR­2 pathway in the duodenum. Furthermore, treatment with SNS restored the normal expression levels of ZO­1, JAM­1 and ß­catenin in FD rats. These findings suggested that the therapeutic effects of SNS on FD were achieved by restoring mucosal barrier integrity and suppressing low­grade inflammation in the duodenum, which was at least partially mediated via the CRF signaling pathway.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Duodenum/drug effects , Duodenum/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction , Animals , Central Nervous System/metabolism , Disease Models, Animal , Duodenum/pathology , Dyspepsia , Enteritis/diagnosis , Enteritis/drug therapy , Enteritis/etiology , Enteritis/metabolism , Gene Expression , Immunohistochemistry , Intestinal Mucosa/pathology , Male , RNA, Messenger/genetics , Rats
12.
BMC Complement Altern Med ; 19(1): 147, 2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31234859

ABSTRACT

BACKGROUND: Ethanol withdrawal (EtOHW) anxiety is a crucial risk factor for alcoholic relapse. The neuropeptide nociceptin/orphanin FQ (N/OFQ) acts upon its receptor (NOP) to antagonize corticotropin-releasing factor (CRF) and elicit anxiolytic actions. Semen Ziziphi Spinosae (SZS), a prototypical hypnotic-sedative herb in Oriental medicine, exhibits anxiolytic effects during nicotine withdrawal by improving amygdaloid CRF/CRF1 receptor (CRFR1) signaling. Therefore, we evaluated the effects of SZS on EtOHW anxiety and the involvement of amygdaloid CRF/CRFR1 and N/OFQ/NOP pathways. METHODS: Male Sprague Dawley rats received intraperitoneal injections of 2 g/kg EtOH (20% v/v) once daily for 28 d followed by a 3-d withdrawal. During EtOHW, the rats were given once-daily intragastric treatments of a methanol extract of SZS (MESZS, 60 or 180 mg/kg/d). Anxiety-like behaviors were measured with the open field (OF) and elevated plus maze (EPM) tests, and plasma corticosterone (CORT) levels were examined by an enzyme-linked immunosorbent assay. mRNA and protein expression levels of the neuropeptides and their receptors were determined by quantitative polymerase chain reaction and Western blot assays. RESULTS: MESZS increased the distance traveled in the center zone of the OF and dose-dependently elongated the duration of staying in the center zone in EtOHW rats. MESZS increased both the number of entries into and the time spent in the open arms of the EPM by EtOHW rats. And, MESZS inhibited the over secretion of plasma CORT during EtOHW. EtOHW enhanced CRF and CRFR1 gene and protein expression in the central nucleus of the amygdala (CeA), which were inhibited by 180 mg/kg/d MESZS. EtOHW increased amygdaloid NOP mRNA and protein expression but spared N/OFQ mRNA expression, and 180 mg/kg/d MESZS further promoted these increases. Additionally, a post-MESZS intra-CeA infusion of either CRF or the selective NOP antagonist UFP-101 abolished the expected anxiolytic effect of 180 mg/kg/d MESZS. CONCLUSIONS: These results suggest that MESZS ameliorates EtOHW anxiety by improving both CRF/CRFR1 and N/OFQ/NOP transmissions in the CeA.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Anxiety/drug therapy , Central Amygdaloid Nucleus/drug effects , Ethanol/adverse effects , Neuropeptides/metabolism , Substance Withdrawal Syndrome/complications , Ziziphus/chemistry , Animals , Anxiety/etiology , Anxiety/genetics , Anxiety/metabolism , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
13.
Physiol Behav ; 199: 210-218, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30472394

ABSTRACT

Oxytocin (OXT) is known to influence on social behaviors, including intermale aggression and hypothalamic-pituitary-adrenal (HPA) axis activity. However, there are no data on the effects of oxytocin on intermale aggression and HPA axis activity in rats selected for elimination and enhancement of aggressiveness towards humans. The aim of this study is to elucidate the role of oxytocin in expression of aggressive behavior and stress response in Norway rats selected for elimination (tame) and enhancement (aggressive) of an aggressive-defensive reaction to humans. Oxytocin was administered to males via nasal applications once or for 5 days (daily). Resident-intruder test showed that in aggressive males, single oxytocin administration caused an increase in the latent period of aggressive interactions and a decrease in the percentage of direct aggression time (not including the time of lateral threat postures) as compared to the control aggressive rats administered with saline. After a 5-day oxytocin administration, aggressive animals demonstrated shorter time of aggressive interactions compared to the control rats. Resident-intruder test revealed no significant changes in behavior of tame rats after single oxytocin administration, while multiple administration caused an increase in aggressive behavior in tame rats. Oxytocin applications caused an elevation of corticosterone level after restriction in aggressive males, but did not affect expression of Crh, Crh1 and Crhr2 genes in hypothalamus in either tame or aggressive rats. The data obtained indicate significant role of oxytocinergic system in the behavior formed in the process of selection by reaction to humans.


Subject(s)
Aggression/drug effects , Behavior, Animal/drug effects , Corticosterone/blood , Oxytocin/administration & dosage , Administration, Intranasal , Aggression/physiology , Animals , Behavior, Animal/physiology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Rats , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Social Behavior
14.
Int J Mol Sci ; 19(11)2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30400578

ABSTRACT

In our previous investigation, we found that agarwood essential oil (AEO) has a sedative-hypnotic effect. Sedative-hypnotic drugs usually have an anxiolytic effect, where concomitant anxiety and depression are a common comorbidity. Therefore, this study further investigated the anxiolytic and antidepressant effects of AEO using a series of animal behavior tests on a restraint stress-induced mice model. The elevated plus maze (EPM) test, the light dark exploration (LDE) test, and the open field (OF) test demonstrated that AEO has a significant anxiolytic effect. Simultaneously, the tail suspension (TS) test and the forced swimming (FS) test illuminated that AEO has an antidepressant effect with the immobility time decreased. Stress can cause cytokine and nitric oxide (NO) elevation, and further lead to hypothalamic-pituitary-adrenal (HPA) axis hyperactivity. AEO was shown to dose-dependently inhibit the levels of cytokines, including interleukin 1α (IL-1α), IL-1ß, and IL-6 in serum, significantly decrease the mRNA level of neural nitric oxide synthase (nNOS) in the cerebral cortex and hippocampus, and inhibit the nNOS protein level in the hippocampus. Concomitant measurements of the HPA axis upstream regulator corticotropin releasing factor (CRF) and its receptor CRFR found that AEO significantly decreases the gene expression of CRF, and significantly inhibits the gene transcription and protein expression of CRFR in the cerebral cortex and hippocampus. Additionally, AEO dose-dependently reduces the concentrations of adrenocorticotropic hormone (ACTH) and corticosterone (CORT) downstream of the HPA axis, as measured by ELISA kits. These results together demonstrate that AEO exerts anxiolytic and antidepressant effects which are related to the inhibition of CRF and hyperactivity of the HPA axis.


Subject(s)
Anxiety/drug therapy , Depression/drug therapy , Hypothalamo-Hypophyseal System/pathology , Oils, Volatile/therapeutic use , Pituitary-Adrenal System/pathology , Restraint, Physical , Stress, Physiological , Thymelaeaceae/chemistry , Adrenocorticotropic Hormone/blood , Animals , Anxiety/blood , Anxiety/etiology , Brain/drug effects , Brain/enzymology , Brain/pathology , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Cytokines/blood , Darkness , Depression/blood , Depression/etiology , Hindlimb Suspension , Hypothalamo-Hypophyseal System/drug effects , Inflammation Mediators/metabolism , Male , Maze Learning/drug effects , Mice, Inbred ICR , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Oils, Volatile/pharmacology , Pituitary-Adrenal System/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Swimming , Transcription, Genetic/drug effects
15.
PLoS One ; 13(9): e0203704, 2018.
Article in English | MEDLINE | ID: mdl-30192883

ABSTRACT

Functional gastrointestinal disorders (FGIDs) are characterized by dysregulated gut-brain interactions. Emerging evidence shows that low-grade mucosal inflammation and immune activation contribute to FGIDs, including functional dyspepsia (FD). Stress plays an important role in the onset of FD symptoms. In human subjects with FD, presence of gastric mast cells has been reported, but factors that influence mast cell infiltration remain uncharacterized. Corticotropin-releasing factor (CRF) initiates the body's stress response and is known to degranulate mast cells. In this study, we delineated the role of the CRF system in the pathogenesis of FD in a rat model. Gastric irritation in neonate rat pups with iodoacetamide (IA) was used to induce FD-like symptoms. RNA interference (RNAi) was used to silence gastric CRF expression. Mast cell infiltrate in the stomach increased by 54% in IA-treated rats compared to controls and CRF-RNAi tended to decrease gastric mast cell infiltrate. Sucrose intake decreased in IA-treated rats and mast cell numbers showed a negative association with sucrose intake. IA treatment and transient silencing of gastric CRF increased hypothalamic CRF levels. In IA-treated rats, gastric levels of CRF receptor 2 (CRF2) decreased by ~76%, whereas hypothalamic CRF receptor 1 (CRF1) levels increased. Plasma levels of TNF-α showed a positive correlation with plasma CRF levels. Levels of phosphorylated p38 and ERK1/2 in the stomach showed a positive correlation with gastric CRF levels. Thus, CRF may contribute to low grade inflammation via modulating mast cell infiltration, cytokine levels, MAPK signaling, and the gut-brain axis.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Dyspepsia/immunology , Dyspepsia/metabolism , Gastric Mucosa/metabolism , Mast Cells/cytology , Animals , Behavior, Animal , Cell Count , Corticotropin-Releasing Hormone/deficiency , Corticotropin-Releasing Hormone/genetics , Disease Models, Animal , Dyspepsia/pathology , Dyspepsia/physiopathology , Gastric Mucosa/drug effects , Gastrointestinal Transit/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Iodoacetamide/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mast Cells/drug effects , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
World J Gastroenterol ; 24(16): 1766-1778, 2018 Apr 28.
Article in English | MEDLINE | ID: mdl-29713130

ABSTRACT

AIM: To explore the significance of corticotropin-releasing hormone (CRH)-receptor (R)2 in mucosal healing of dextran sulfate sodium (DSS)-induced colitis and the effect of Tong-Xie-Yao-Fang (TXYF) on CRH-R2 expression and regulation. METHODS: Ulcerative colitis was induced in mice by administration of 3% (w/v) DSS for 7 d. Once the model was established, mice were administered urocortin-2 (30 µg/kg), a peptide which binds exclusively to CRH-R2, or various doses of aqueous TXYF extracts (2.8-11.2 g/kg), a CRH-R2 antagonist Astressin (Ast)2B (20 µg/kg), Ast2B + Ucn2, or Ast2B with various doses of aqueous TXYF extracts for 9 d. Colonic mucosal permeability was then evaluated by measuring the fluorescence intensity in serum. The colitis disease activity index (DAI), histology, body weight loss and colon length were assessed to evaluate the condition of colitis. Terminal deoxynucleotidyl transferase dUTP nick-end labeling was used to detect apoptosis of the intestinal epithelial cells. The expression level of Ki-67 represented the proliferation of colonic epithelial cells and was detected by immunohistochemistry. The expression levels of inflammation cytokines IL-6, TNF-α and CXCL-1 were examined in colon tissues using real-time PCR and ELISA kits. RESULTS: Compared with the DSS group, mice treated with the CRH-R2 antagonist Ast2B showed greater loss of body weight, shorter colon lengths (4.90 ± 0.32 vs 6.21 ± 0.34 cm, P < 0.05), and higher DAI (3.61 ± 0.53 vs 2.42 ± 0.32, P < 0.05) and histological scores (11.50 ± 1.05 vs 8.33 ± 1.03, P < 0.05). Additionally, the Ast2B group showed increased intestinal permeability (2.76 ± 0.11 µg/mL vs 1.47 ± 0.11 µg/mL, P < 0.001), improved secretion of inflammatory cytokines in colon tissue, and reduced colonic epithelial cell proliferation (4.97 ± 4.25 vs 22.51 ± 8.22, P < 0.05). Increased apoptosis (1422.39 ± 90.71 vs 983.01 ± 98.17, P < 0.001) was also demonstrated. The Ucn2 group demonstrated lower DAI (0.87 ± 0.55 vs 2.42 ± 0.32, P < 0.001) and histological scores (4.33 ± 1.50 vs 8.33 ± 1.03, P < 0.05). Diminished weight loss, longer colon length (9.58 ± 0.62 vs 6.21 ± 0.34 cm, P < 0.001), reduced intestinal permeability (0.75 ± 0.07 vs 1.47 ± 0.11 µg/mL, P < 0.001), inhibited secretion of inflammatory cytokines in colon tissue and increased colonic epithelial cell proliferation (90.04 ± 15.50 vs 22.51 ± 8.22, P < 0.01) were all observed. Reduced apoptosis (149.55 ± 21.68 vs 983.01 ± 98.17, P < 0.05) was also observed. However, significant statistical differences in the results of the Ast2B group and Ast2B + Ucn2 group were observed. TXYF was also found to ameliorate symptoms of DSS-induced colitis in mice and to promote mucosal repair like Ucn2. There were significant differences between the Ast2B + TXYF groups and the TXYF groups. CONCLUSION: CRH-R2 activates the intestinal mucosal antiinflammatory response by regulating migration, proliferation and apoptosis of intestinal epithelial cells in colitis-induced mice, and plays an important antiinflammatory role. TXYF promotes mucosal repair in colitis mice by regulating CRH-R2.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Colon/drug effects , Dextran Sulfate , Drugs, Chinese Herbal/pharmacology , Intestinal Mucosa/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Wound Healing/drug effects , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokine CXCL1/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ki-67 Antigen/metabolism , Male , Mice, Inbred ICR , Permeability , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
17.
Int J Cancer ; 142(2): 334-346, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28929494

ABSTRACT

Colorectal cancer (CRC) responds poorly to immuno-mediated cytotoxicity. Underexpression of corticotropin-releasing-hormone-receptor-2 (CRHR2) in CRC, promotes tumor survival, growth and Epithelial to Mesenchymal Transition (EMT), in vitro and in vivo. We explored the role of CRHR2 downregulation in CRC cell resistance to Fas/FasL-mediated apoptosis and the underlying molecular mechanism. CRC cell sensitivity to CH11-induced apoptosis was compared between Urocortin-2 (Ucn2)-stimulated parental and CRHR2-overexpressing CRC cell lines and targets of CRHR2/Ucn2 signaling were identified through in vitro and ex vivo analyses. Induced CRHR2/Ucn2 signaling in SW620 and DLD1 cells increased specifically their sensitivity to CH11-mediated apoptosis, via Fas mRNA and protein upregulation. CRC compared to control tissues had reduced Fas expression that was associated with lost CRHR2 mRNA, poor tumor differentiation and high risk for distant metastasis. YY1 silencing increased Fas promoter activity in SW620 and re-sensitized them to CH11-apoptosis, thus suggesting YY1 as a putative transcriptional repressor of Fas in CRC. An inverse correlation between Fas and YY1 expression was confirmed in CRC tissue arrays, while elevated YY1 mRNA was clinically relevant with advanced CRC grade and higher risk for distant metastasis. CRHR2/Ucn2 signaling downregulated specifically YY1 expression through miR-7 elevation, while miR-7 modulation in miR-7high SW620-CRHR2+ and miR-7low HCT116 cells, had opposite effects on YY1 and Fas expressions and cell sensitivity to CH11-killing. CRHR2/Ucn2 signaling is a negative regulator of CRC cell resistance to Fas/FasL-apoptosis via targeting the miR-7/YY1/Fas circuitry. CRHR2 restoration might prove effective in managing CRC response to immune-mediated apoptotic stimuli.


Subject(s)
Apoptosis , Colorectal Neoplasms/pathology , Corticotropin-Releasing Hormone/metabolism , MicroRNAs/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/metabolism , YY1 Transcription Factor/metabolism , fas Receptor/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Corticotropin-Releasing Hormone/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Receptors, Corticotropin-Releasing Hormone/genetics , Signal Transduction , Tumor Cells, Cultured , Urocortins/genetics , YY1 Transcription Factor/genetics , fas Receptor/genetics
18.
Gen Comp Endocrinol ; 258: 91-98, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28774755

ABSTRACT

It is well established that hypothalamic neurons producing the peptide corticotropin-releasing factor (CRF) play a key role in stress adaptation, including reduction of food intake when a threat or stressor is present. We have previously reported on the presence of an intrinsic CRF signaling system within the optic tectum (OT), a brain area that plays a key role in visually guided prey capture/predator avoidance decisions. To better understand the potential role of tectal CRF neurons in regulating adaptive behavior and energy balance during stress we examined evidence for modulation of tectal CRF neuronal activity after stressor exposure and food deprivation in the African clawed frog Xenopus laevis. We tested two predictions, 1) that exposure to categorically distinct stressors (ether vapors and shaking) will reduce food intake and modulate the activity of tectal CRF cells, and 2) that food deprivation will modulate the activity of tectal CRF cells. Exposure to ether increased tectal content of CRF and CRF transcript, but lowed CRFR1 transcript abundance. Two weeks of food deprivation reduced total fat stores in frogs and decreased tectal content of CRF content while having no effect on CRF and CRFR1 transcript abundance. Our data are consistent with a role for tectal CRF neurons in modulating food intake in response to certain stressors.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Fasting , Neurons/metabolism , Stress, Physiological , Superior Colliculi/metabolism , Xenopus laevis/metabolism , Adaptation, Physiological , Animals , Eating , Food Deprivation , Hypothalamus/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Xenopus laevis/genetics , Xenopus laevis/physiology
19.
Reprod Sci ; 25(9): 1336-1348, 2018 09.
Article in English | MEDLINE | ID: mdl-29137551

ABSTRACT

Treatments for endometriosis include pharmacological or surgical procedures that produce significant side effects. We aimed to determine how environmental enrichment (EE) could impact the progression of endometriosis using the autotransplantation rat model. Female rats were exposed to EE (endo-EE: toys and nesting materials, 4 rats per cage, larger area enclosure) or no enrichment (endo-NE: 2 rats per cage) starting on postnatal day 21. After 8 weeks, sham surgery or surgical endometriosis was induced by suturing uterine horn tissue next to the intestinal mesentery, then allowed to progress for 60 days during which EE or NE continued. At the time of killing, we measured anxiety behaviors, collected endometriotic vesicles and uterus, and processed for quantitative real-time polymerase chain reaction for corticotropin-releasing hormone (CRH), urocortin-1, CRH receptors type 1 and type 2, and glucocorticoid receptor (GR). Endometriosis did not affect anxiety-like behaviors, yet rats in enriched conditions showed lower basal anxiety behaviors than the nonenriched group. Importantly, the endo-EE group showed a 28% reduction in the number of endometriosis vesicles and the vesicles were significantly smaller compared to the endo-NE group. Endometriosis increased CRH and GR only in the vesicles of endo-NE, and this increase was dampened in the endo-EE. However, urocortin 1 was increased in the vesicles of the endo-EE group, suggesting different pathways of activation of CRH receptors in this group. Our results suggest that the use of multimodal complementary therapies that reduce stress in endometriosis could be an effective and safe treatment alternative, with minimal side effects.


Subject(s)
Behavior, Animal/physiology , Endometriosis/therapy , Environment , Housing, Animal , Uterus/metabolism , Animals , Anxiety/metabolism , Anxiety/psychology , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Disease Progression , Endometriosis/metabolism , Endometriosis/psychology , Female , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/metabolism , Stress, Psychological/metabolism , Stress, Psychological/psychology , Urocortins/metabolism
20.
Nutr Neurosci ; 21(1): 49-58, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27686011

ABSTRACT

OBJECTIVE: The objective was to determine the effects of high-protein and high-fat diets, and fasting and refeeding, on appetite regulation in chicks. METHODS: Day of hatch chicks were fed one of four diets: basal, high protein (25% crude protein), and 15 and 30% high fat (15 and 30% metabolizable energy derived from soybean oil, respectively), and assigned to one of three treatments at 4 days: (1) access to feed, (2) 3 hours of fasting, or (3) fasting followed by 1 hour of refeeding. The hypothalamus was collected, total RNA isolated, and mRNA abundance measured. RESULTS: Food intake was reduced in chicks fed the high-protein and high-fat diets. Agouti-related peptide, neuropeptide Y (NPY), NPY receptors 1, 2, and 5, melanocortin receptors 3 and 4 (MC3R and 4R, respectively), mesotocin, corticotropin-releasing factor (CRF), and CRF receptor sub-type 2 (CRFR2) mRNAs were greatest in chicks that consumed the basal diet. Refeeding was associated with increased MC3R mRNA in the high-protein diet group. CRFR2 mRNA was increased by fasting and refeeding in chicks that consumed the high-protein diet. DISCUSSION: Food intake and hypothalamic gene expression of some important appetite-associated factors were reduced in chicks fed the high-protein or high-fat diets. Fasting and refeeding accentuated several differences and results suggest that the CRF and melanocortin pathways are involved.


Subject(s)
Appetite Regulation , Diet, High-Fat , Diet, High-Protein , Hypothalamus/physiology , Animals , Appetite , Body Weight , Chickens , Fasting , Gene Expression Regulation , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL