Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Sci Rep ; 10(1): 18025, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093538

ABSTRACT

Sleep disturbances are commonly found in trauma-exposed populations. Additionally, trauma exposure results in fear-associated memory impairments. Given the interactions of sleep with learning and memory, we hypothesized that increasing sleep duration following trauma exposure would restore overall function and improve trauma-induced fear-associated memory dysfunction. Here, we utilized single prolonged stress, a validated rodent model of post-traumatic stress disorder, in combination with optogenetic activation of hypothalamic melanin-concentrating hormone containing cells to increase sleep duration. The goal of this work was to ascertain if post-trauma sleep increases are sufficient to improve fear-associated memory function. In our laboratory, optogenetic stimulation after trauma exposure was sufficient to increase REM sleep duration during both the Light and Dark Phase, whereas NREM sleep duration was only increased during the Dark Phase of the circadian day. Interestingly though, animals that received optogenetic stimulation showed significantly improved fear-associated memory processing compared to non-stimulated controls. These results suggest that sleep therapeutics immediately following trauma exposure may be beneficial and that post-trauma sleep needs to be further examined in the context of the development of post-traumatic stress disorder.


Subject(s)
Fear , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Memory Disorders/prevention & control , Optogenetics , Pituitary Hormones/administration & dosage , Sleep Wake Disorders/prevention & control , Sleep, REM , Stress Disorders, Post-Traumatic/complications , Animals , Male , Memory Consolidation , Memory Disorders/etiology , Rats , Rats, Long-Evans , Sleep Wake Disorders/etiology
2.
Article in English | MEDLINE | ID: mdl-32328034

ABSTRACT

RF amide-related peptide 3 (RFRP-3) is a reproductive inhibitor and an endogenous orexigenic neuropeptide that may be involved in energy homeostasis. In this study, we evaluated the effect of acute or chronic RFRP-3 treatment (administered via intraperitoneal injection) on the food intake, meal microstructure and weight of rats, as well as the mechanism through which RFRP-3 is involved in glucose metabolism in the pancreas and glucose disposal tissues of rat in vivo. Our results showed that the intraperitoneal administration of RFRP-3 to rats resulted in marked body mass increased, hyperphagia, hyperlipidemia, hyperglycemia, glucose intolerance, hypoinsulinism, hyperglucagon, and insulin resistance, as well as significant increases in the size of pancreatic islets and the inflammatory reaction. Thus, we strongly assert that RFRP-3 as a novel neuroendocrine regulator involved in blood glucose homeostasis.


Subject(s)
Appetite Regulation/drug effects , Carbohydrate Metabolism/drug effects , Glucose/metabolism , Hypothalamic Hormones/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Female , Glucose Intolerance/chemically induced , Glucose Intolerance/metabolism , Homeostasis/drug effects , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/physiology , Inflammation/chemically induced , Inflammation/metabolism , Injections, Intraperitoneal , Insulin Resistance , Male , Obesity/chemically induced , Obesity/metabolism , Rats , Rats, Sprague-Dawley
3.
J Psychopharmacol ; 34(4): 478-489, 2020 04.
Article in English | MEDLINE | ID: mdl-31909693

ABSTRACT

BACKGROUND: Identifying neural substrates that are differentially affected by drugs of abuse and natural rewards is key to finding a target for an efficacious treatment for substance abuse. Melanin-concentrating hormone is a polypeptide with an inhibitory effect on the mesolimbic dopamine system. Here we test the hypothesis that melanin-concentrating hormone in the lateral hypothalamus and nucleus accumbens shell is differentially involved in the regulation of morphine and food-rewarded behaviors. METHODS: Male Sprague-Dawley rats were trained with morphine (5.0 mg/kg, subcutaneously) or food pellets (standard chow, 10-14 g) to induce a conditioned place preference, immediately followed by extinction training. Melanin-concentrating hormone (1.0 µg/side) or saline was infused into the nucleus accumbens shell or lateral hypothalamus before the reinstatement primed by morphine or food, and locomotor activity was simultaneously monitored. As the comparison, melanin-concentrating hormone was also microinjected into the nucleus accumbens shell or lateral hypothalamus before the expression of food or morphine-induced conditioned place preference. RESULTS: Microinfusion of melanin-concentrating hormone into the nucleus accumbens shell (but not into the lateral hypothalamus) prevented the reinstatement of morphine conditioned place preference but had no effect on the reinstatement of food conditioned place preference. In contrast, microinfusion of melanin-concentrating hormone into the lateral hypothalamus (but not in the nucleus accumbens shell) inhibited the reinstatement of food conditioned place preference but had no effect on the reinstatement of morphine conditioned place preference. CONCLUSIONS: These results suggest a clear double dissociation of melanin-concentrating hormone in morphine/food rewarding behaviors and melanin-concentrating hormone in the nucleus accumbens shell. Melanin-concentrating hormone could be a potential target for therapeutic intervention for morphine abuse without affecting natural rewards.


Subject(s)
Drug-Seeking Behavior/drug effects , Feeding Behavior/drug effects , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/pharmacology , Melanins/pharmacology , Morphine/pharmacology , Nucleus Accumbens/metabolism , Pituitary Hormones/pharmacology , Animals , Conditioning, Operant/drug effects , Hypothalamic Hormones/administration & dosage , Male , Melanins/administration & dosage , Microinjections , Pituitary Hormones/administration & dosage , Rats , Rats, Sprague-Dawley , Reward
4.
Anim Biotechnol ; 31(4): 335-349, 2020 Aug.
Article in English | MEDLINE | ID: mdl-30961416

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH) plays an important role in reproduction by inhibiting the expression of gonadotropins in birds and mammals, but in fishes, it is ambiguous. In this study, we cloned 606 bp long cDNA of GnIH from Catla catla brain (cGnIH). The encoded preproGnIH peptide generated three putative peptides (cGnIH-I, -II, -III) of different size. Phylogenetic analysis of GnIH showed clustering of different peptide sequence with its orthologs in separate clades. The real-time PCR analysis showed the expression of cGnIH in brain, gonads, intestine, stomach, heart, gill and liver with the highest expression in the brain and gonads of both sexes. The basal GnIH mRNA expression was higher in spawning and spent phase of the male brain and spawning phase of the female brain. In testis, the expression was highest in spent phase, while in ovary the expression did not change significantly during reproductive phases. The in vivo experiment of cGnIH-III peptide exhibited the higher expression of HPG axis genes, lhb, fshb, cgnrh, kiss2 and kiss1r and serum hormone level of LH and FSH as soon as 3 h after the intramuscular delivery. These results suggest that the GnIH is positively involved in regulation of reproduction in HPG axis of C. catla.


Subject(s)
Cyprinidae/genetics , Cyprinidae/physiology , Fish Proteins , Hypothalamic Hormones , Reproduction/drug effects , Amino Acid Sequence , Animals , Brain/drug effects , Brain/metabolism , Female , Fish Proteins/administration & dosage , Fish Proteins/chemistry , Fish Proteins/pharmacology , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/chemistry , Hypothalamic Hormones/pharmacology , Injections, Intramuscular , Male , Peptides/administration & dosage , Peptides/chemistry , Peptides/pharmacology , Pituitary Hormones/blood , Testis/drug effects , Testis/metabolism
5.
Mol Neurobiol ; 56(12): 8076-8086, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31183806

ABSTRACT

Melanin-concentrating hormone (MCH) is a highly conserved neuropeptide known to exhibit important functions in the brain. Some studies have reported that MCH improves memory by promoting memory retention. However, the precise molecular mechanisms by which MCH enhances memory impairment have yet to be fully elucidated. In this study, MCH was administered to the scopolamine-induced memory-impaired mice via the nasal cavity to examine the acute effects of MCH and Alzheimer's disease (AD) mouse models to evaluate the chronic effects of MCH. MCH improved memory impairment in both models and reduced soluble amyloid beta in the cerebral cortex of APP/PS1 transgenic mice. In vitro assays also showed that MCH inhibits amyloid beta-induced cytotoxicity. Furthermore, MCH increased long-term potentiation (LTP) in the hippocampus of wild-type and 5XFAD AD mouse model. To further elucidate the mechanisms of the chronic effect of MCH, the levels of phosphorylated CREB and GSK3ß, and the expression of BDNF, TrkB and PSD95 were examined in the cerebral cortex and hippocampus. Our findings indicate that MCH might have neuroprotective effects via downstream pathways associated with the enhancement of neuronal synapses and LTP. This suggests a therapeutic potential of MCH for the treatment of neurodegenerative diseases such as AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Disease Models, Animal , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Memory Disorders/drug therapy , Memory Disorders/metabolism , Pituitary Hormones/administration & dosage , Administration, Intranasal , Animals , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Female , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nasal Cavity/drug effects , Nasal Cavity/metabolism , Pregnancy
6.
Neuropeptides ; 74: 70-81, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30642579

ABSTRACT

Melanin-concentrating hormone (MCH) is a neuropeptide present in neurons located in the hypothalamus that densely innervate serotonergic cells in the dorsal raphe nucleus (DRN). MCH administration into the DRN induces a depressive-like effect through a serotonergic mechanism. To further understand the interaction between MCH and serotonin, we used primary cultured serotonergic neurons to evaluate the effect of MCH on serotonergic release and metabolism by HPLC-ED measurement of serotonin (5-HT) and 5-hydroxyindolacetic acid (5-HIAA) levels. We confirmed the presence of serotonergic neurons in the E14 rat rhombencephalon by immunohistochemistry and showed for the first time evidence of MCHergic fibers reaching the area. Cultures obtained from rhombencephalic tissue presented 2.2 ±â€¯0.7% of serotonergic and 48.9 ±â€¯5.4% of GABAergic neurons. Despite the low concentration of serotonergic neurons, we were able to measure basal cellular and extracellular levels of 5-HT and 5-HIAA without the addition of any serotonergic-enhancer drug. As expected, 5-HT release was calcium-dependent and induced by depolarization. 5-HT extracellular levels were significantly increased by incubation with serotonin reuptake inhibitors (citalopram and nortriptyline) and a monoamine-oxidase inhibitor (clorgyline), and were not significantly modified by a 5-HT1A autoreceptor agonist (8-OHDPAT). Even though serotonergic cells responded as expected to these pharmacological treatments, MCH did not induce significant modifications of 5-HT and 5-HIAA extracellular levels in the cultures. Despite this unexpected result, we consider that assessment of 5-HT and 5-HIAA levels in primary serotonergic cultures may be an adequate approach to study the effect of other drugs and modulators on serotonin release, uptake and turnover.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Raphe Nuclei/metabolism , Serotonin/metabolism , Animals , GABAergic Neurons/cytology , Hypothalamic Hormones/administration & dosage , Hypothalamus/cytology , Melanins/administration & dosage , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/cytology , Neurons/drug effects , Pituitary Hormones/administration & dosage , Primary Cell Culture , Raphe Nuclei/cytology , Raphe Nuclei/drug effects , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism
7.
Article in English | MEDLINE | ID: mdl-30114526

ABSTRACT

Phoenixin (Pnx) is an endogenous peptide known to be involved in reproduction and food intake in rats, with two active isoforms, phoenixin-14 (Pnx-14) and phoenixin-20 (Pnx-20). However, little is known about the functions of Pnx in teleost. Here, pnx was cloned and was detected in all tissues of both male and female in spotted scat (Scatophagus argus), including growth axis, hypothalamus, pituitary, and liver. Real-time PCR analysis showed that pnx in the hypothalamus increased significantly after 2 d and 7 d fasting, while reduced significantly after re-feeding (P < 0.05). When pituitary and liver fragments were cultured in vitro with Pnx-14 and Pnx-20 (10 nM and 100 nM) for 6 h, the expression of ghrhr (growth hormone-releasing hormone receptor) and gh (growth hormone) in the pituitary, and ghr1 (growth hormone receptor 1) in the liver increased significantly, except ghr2 (growth hormone receptor 2) incubated with 10 nM and 100 nM Pnx-20 and ghr1 incubated with 10 nM Pnx-20. Similarly, the expression of ghrhr and gh in the pituitary, as well as ghr1 and ghr2 in the liver, increased significantly after injecting S. argus with Pnx-14 and Pnx-20 (10 ng/g and 100 ng/g body weight). These results indicate that Pnx is likely to be involved in the regulation of food intake, and also regulates the growth of S. argus by increasing ghrhr and gh expression in the pituitary, ghr1 and ghr2 in the liver, and ghr1 directly in the liver.


Subject(s)
Energy Intake , Fish Proteins/metabolism , Gene Expression Regulation, Developmental , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Peptide Hormones/metabolism , Perciformes/physiology , Animals , Aquaculture , China , Energy Intake/drug effects , Female , Fish Proteins/administration & dosage , Fish Proteins/genetics , Fish Proteins/pharmacology , Gene Expression Regulation, Developmental/drug effects , Growth Hormone/agonists , Growth Hormone/genetics , Growth Hormone/metabolism , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/genetics , Hypothalamic Hormones/pharmacology , Hypothalamus/drug effects , Injections, Intraperitoneal , Liver/drug effects , Liver/metabolism , Male , Organ Specificity , Peptide Hormones/administration & dosage , Peptide Hormones/genetics , Peptide Hormones/pharmacology , Perciformes/growth & development , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Protein Isoforms/administration & dosage , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Random Allocation , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Somatotropin/agonists , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tissue Culture Techniques/veterinary , Weight Gain
8.
Acta Pharmacol Sin ; 39(5): 770-773, 2018 May.
Article in English | MEDLINE | ID: mdl-29542680

ABSTRACT

In this brief review we summarize the current fndings relative to the discovery of a small peptide ligand, phoenixin (PNX). Using a bioinformatic approach, two novel peptides PNX-14 and PNX-20 containing 14 and 20 amino acids, respectively, were isolated from diverse tissues including the brain, heart, lung and stomach. Mass spectrometry analysis identified a major and minor peak corresponding to PNX-14 and PNX-20, in rat or mouse spinal cord extracts. With the use of a rabbit polyclonal antiserum, phoenixin immunoreactivity (irPNX) was detected in discrete areas of the rodent brain including several hypothalamic subnuclei and dorsal motor nucleus of the vagus. In addition, irPNX was detected in a population of sensory ganglion cells including dorsal root ganglion, nodose ganglion and trigeminal ganglion, and in cell processes densely distributed to the superficial layers of the dorsal horn, nucleus of the solitary tract and spinal trigeminal tract. irPNX cell processes were also detected in the skin and myenteric plexus, suggesting a brain-gut and/or brain-skin connection. Pharmacological studies show that PNX-14 injected subcutaneously to the nape of the neck of mice provoked dose-dependent repetitive scratching bouts directed to the back of the neck with the hindpaws. Our result suggests that the peptide PNX-14 and/or PNX-20, may serve as one of the endogenous signal molecules transducing itch sensation. Additionally, results from other laboratories show that exogenous PNX may affect a number of diverse behaviors such as memory formation, depression, reproduction, food-intake and anxiolytic-like behaviors.


Subject(s)
Hypothalamic Hormones/physiology , Peptide Hormones/physiology , Peptides/physiology , Amino Acid Sequence , Animals , Humans , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/chemistry , Hypothalamus/metabolism , Memory/physiology , Myenteric Plexus/metabolism , Peptide Hormones/administration & dosage , Peptide Hormones/chemistry , Peptides/administration & dosage , Peptides/chemistry , Pruritus/metabolism , Spinal Cord/metabolism
9.
Sci Rep ; 8(1): 707, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335480

ABSTRACT

The melanin-concentrating hormone (MCH) is a peptidergic neuromodulator synthesized by neurons in the lateral hypothalamus and zona incerta. MCHergic neurons project throughout the central nervous system, indicating the involvements of many physiological functions, but the role in pain has yet to be determined. In this study, we found that pMCH-/- mice showed lower baseline pain thresholds to mechanical and thermal stimuli than did pMCH+/+ mice, and the time to reach the maximum hyperalgesic response was also significantly earlier in both inflammatory and neuropathic pain. To examine its pharmacological properties, MCH was administered intranasally into mice, and results indicated that MCH treatment significantly increased mechanical and thermal pain thresholds in both pain models. Antagonist challenges with naltrexone (opioid receptor antagonist) and AM251 (cannabinoid 1 receptor antagonist) reversed the analgesic effects of MCH in both pain models, suggesting the involvement of opioid and cannabinoid systems. MCH treatment also increased the expression and activation of CB1R in the medial prefrontal cortex and dorsolateral- and ventrolateral periaqueductal grey. The MCH1R antagonist abolished the effects induced by MCH. This is the first study to suggest novel analgesic actions of MCH, which holds great promise for the application of MCH in the therapy of pain-related diseases.


Subject(s)
Analgesics/pharmacology , Hypothalamic Hormones/pharmacology , Melanins/pharmacology , Pain Perception/drug effects , Pituitary Hormones/pharmacology , Administration, Intranasal , Analgesics/administration & dosage , Animals , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/deficiency , Melanins/administration & dosage , Melanins/deficiency , Mice , Mice, Knockout , Pituitary Hormones/administration & dosage , Pituitary Hormones/deficiency
10.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R623-R628, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29364701

ABSTRACT

The newly described hypothalamic peptide, phoenixin, is produced in the hypothalamus and adenohypophysis, where it acts to control reproductive hormone secretion. Both phoenixin and its receptor GPR173 are expressed in the hypothalamic supraoptic (SON) and paraventricular (PVN) nuclei, suggesting additional, nonreproductive effects of the peptide to control vasopressin (AVP) or oxytocin (OT) secretion. Hypothalamo-neurohypophysial explants released AVP but not OT in response to phoenixin. Intracerebroventricular administration of phoenixin into conscious, unrestrained male and female rats significantly increased circulating AVP, but not OT, levels in plasma, and it increased immediate early gene expression in the supraoptic nuclei of male rats. Bath application of phoenixin in hypothalamic slice preparations resulted in depolarization of PVN neurons, indicating a direct, neural action of phoenixin in the hypothalamus. Our results suggest that the newly described, hypothalamic peptide phoenixin, in addition to its effects on hypothalamic and pituitary mechanisms controlling reproduction, may contribute to the physiological mechanisms regulating fluid and electrolyte homeostasis.


Subject(s)
Arginine Vasopressin/metabolism , Hypothalamic Hormones/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Peptide Hormones/physiology , Animals , Arginine Vasopressin/blood , Female , Gene Expression Regulation , Genes, fos , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/physiology , Hypothalamo-Hypophyseal System/metabolism , In Vitro Techniques , Injections, Intraventricular , Male , Membrane Potentials , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats, Sprague-Dawley , Secretory Pathway/drug effects , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism
11.
Neuropharmacology ; 130: 62-70, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29191753

ABSTRACT

Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.


Subject(s)
Eating/drug effects , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Pituitary Hormones/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/metabolism , Dependovirus , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , MAP Kinase Signaling System/drug effects , Male , Melanins/metabolism , Mice , Mice, Inbred C57BL , Pituitary Hormones/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/metabolism , Ribosomal Protein S6 Kinases/drug effects , Ribosomal Protein S6 Kinases/metabolism
12.
J Med Chem ; 60(22): 9320-9329, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29094944

ABSTRACT

Melanoma is a lethal form of skin cancer. Skin pigmentation, which is regulated by the melanocortin 1 receptor (MC1R), is an effective protection against melanoma. However, the endogenous MC1R agonists lack selectivity for the MC1R and thus can have side effects. The use of noncanonical amino acids in previous MC1R ligand development raises safety concerns. Here we report the development of the first potent and selective hMC1R agonist with only canonical amino acids. Using γ-MSH as a template, we developed a peptide, [Leu3, Leu7, Phe8]-γ-MSH-NH2 (compound 5), which is 16-fold selective for the hMC1R (EC50 = 4.5 nM) versus other melanocortin receptors. Conformational studies revealed a constrained conformation for this linear peptide. Molecular docking demonstrated a hydrophobic binding pocket for the melanocortin 1 receptor. In vivo pigmentation study shows high potency and short duration. [Leu3, Leu7, Phe8]-γ-MSH-NH2 is ideal for inducing short-term skin pigmentation without sun for melanoma prevention.


Subject(s)
Hypothalamic Hormones/pharmacology , Melanocyte-Stimulating Hormones/pharmacology , Receptor, Melanocortin, Type 1/agonists , Skin Pigmentation/drug effects , Animals , Drug Stability , HEK293 Cells , Half-Life , Humans , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/chemical synthesis , Hypothalamic Hormones/pharmacokinetics , Iodine Radioisotopes , Ligands , Melanocyte-Stimulating Hormones/administration & dosage , Melanocyte-Stimulating Hormones/chemical synthesis , Melanocyte-Stimulating Hormones/pharmacokinetics , Molecular Conformation , Molecular Docking Simulation , Receptor, Melanocortin, Type 1/chemistry , Reptiles , alpha-MSH/administration & dosage , alpha-MSH/analogs & derivatives , alpha-MSH/chemical synthesis , alpha-MSH/pharmacokinetics , alpha-MSH/pharmacology
13.
Peptides ; 87: 28-33, 2017 01.
Article in English | MEDLINE | ID: mdl-27845162

ABSTRACT

Neuropeptideglutamic acid-isoleucine (NEI) as well as melanin concentrating hormone (MCH) is cleaved from the 165 amino acid protein, prepro-melanin concentrating hormone (prepro-MCH). Among many physiological roles of MCH, we demonstrated that intracerebroventricular (icv) injection of MCH induced increases in REM sleep episodes as well as in non REM sleep episodes. However, there are no studies on the effect of NEI on the sleep-wake cycle. As for the sites of action of MCH for induction of REM sleep, the ventrolateral periaqueductal gray (vlPAG) has been reported to be one of its site of action. Although MCH neurons contain NEI, GABA, MCH, and other neuropeptides, we do not know which transmitter(s) might induce REM sleep by acting on the vlPAG. Thus, we first examined the effect of icv injection of NEI on the sleep-wake cycle, and investigated how microinjection of either NEI, MCH, or GABA into the vlPAG affected REM sleep in rats. Icv injection of NEI (0.61µg/5µl: n=7) significantly increased the time spent in REM episodes compared to control (saline: 5µl; n=6). Microinjection of either NEI (61ng/0.2µl: n=7), MCH (100ng/0.2µl: n=6) or GABA (250mM/0.2µl: n=7) into the vlPAG significantly increased the time spent in REM episodes and the AUC. Precise hourly analysis of REM sleep also revealed that after those microinjections, NEI and MCH increased REM episodes at the latter phase, compared to GABA which increased REM episodes at the earlier phase. This result suggests that NEI and MCH may induce sustained REM sleep, while GABA may initiate REM sleep. In conclusion, our findings demonstrate that NEI, a cleaved peptide from the same precursor, prepro-MCH, as MCH, induce REM sleep at least in part through acting on the vlPAG.


Subject(s)
Hypothalamic Hormones/metabolism , Melanins/metabolism , Neurons/metabolism , Neuropeptides/administration & dosage , Pituitary Hormones/metabolism , Sleep, REM/drug effects , Animals , Glutamic Acid/administration & dosage , Glutamic Acid/metabolism , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/chemistry , Isoleucine/administration & dosage , Isoleucine/metabolism , Melanins/administration & dosage , Melanins/chemistry , Microinjections , Neurons/drug effects , Neuropeptides/metabolism , Periaqueductal Gray/drug effects , Periaqueductal Gray/metabolism , Periaqueductal Gray/physiology , Pituitary Hormones/administration & dosage , Pituitary Hormones/chemistry , Rats , Sleep, REM/physiology , gamma-Aminobutyric Acid/administration & dosage
14.
Mol Neurobiol ; 54(10): 7706-7721, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27844281

ABSTRACT

Acupuncture has shown the therapeutic effect on various neurodegenerative disorders including Parkinson's disease (PD). While investigating the neuroprotective mechanism of acupuncture, we firstly found the novel function of melanin-concentrating hormone (MCH) as a potent neuroprotective candidate. Here, we explored whether hypothalamic MCH mediates the neuroprotective action of acupuncture. In addition, we aimed at evaluating the neuroprotective effects of MCH and elucidating underlying mechanism in vitro and in vivo PD models. First, we tested whether hypothalamic MCH mediates the neuroprotective effects of acupuncture by challenging MCH-R1 antagonist (i.p.) in mice PD model. We also investigated whether MCH has a beneficial role in dopaminergic neuronal protection in vitro primary midbrain and human neuronal cultures and in vivo MPTP-induced, Pitx3-/-, and A53T mutant mice PD models. Transcriptomics followed by quantitative PCR and western blot analyses were performed to reveal the neuroprotective mechanism of MCH. We first found that hypothalamic MCH biosynthesis was directly activated by acupuncture treatment and that administration of an MCH-R1 antagonist reverses the neuroprotective effects of acupuncture. A novel finding is that MCH showed a beneficial role in dopaminergic neuron protection via downstream pathways related to neuronal survival. This is the first study to suggest the novel neuroprotective action of MCH as well as the involvement of hypothalamic MCH in the acupuncture effects in PD, which holds great promise for the application of MCH in the therapy of neurodegenerative diseases.


Subject(s)
Acupuncture Therapy/methods , Hypothalamic Hormones/biosynthesis , Melanins/biosynthesis , Neuroprotective Agents/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/prevention & control , Pituitary Hormones/biosynthesis , Animals , Cells, Cultured , Humans , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/antagonists & inhibitors , Hypothalamus/metabolism , Male , Melanins/administration & dosage , Melanins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pituitary Hormones/administration & dosage , Pituitary Hormones/antagonists & inhibitors , Treatment Outcome
15.
Neurosci Lett ; 630: 66-69, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27461793

ABSTRACT

A study was performed on the effects of local microinjection of melanin-concentrating hormone (MCH) into the right sublaterodorsal tegmental nucleus (SLD) on sleep and wakefulness in rats prepared for chronic sleep recordings. MCH 200ng significantly decreased rapid-eye-movement sleep (REMS) time during the first and second 2-h of the recording period which was related to the reduction of the number of REMS periods and the increase of REMS latency. It is proposed that REMS inhibition was related to the direct deactivation of SLD glutamatergic neurons by the peptide.


Subject(s)
Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/physiology , Melanins/administration & dosage , Melanins/physiology , Pituitary Hormones/administration & dosage , Pituitary Hormones/physiology , Sleep, REM , Tegmentum Mesencephali/physiology , Wakefulness , Animals , Electroencephalography , Male , Microinjections , Neurons/physiology , Occipital Lobe/physiology , Rats , Rats, Wistar
16.
Brain Res ; 1598: 114-28, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25541366

ABSTRACT

Hypothalamic neurons that utilize melanin-concentrating hormone (MCH) as a neuromodulator are localized in the postero-lateral hypothalamus and incerto-hypothalamic area. These neurons send dense projections to the dorsal raphe nucleus (DRN). Serotonergic neurons of the DRN are involved in the control of sleep and play a critical role in major depression. Previously, we demonstrated that microinjections of MCH into the DRN resulted in an increase in REM sleep and produce a depressive-like effect. In the present study we examined the mechanisms that mediate these effects by employing neuroanatomical and electrophysiological techniques. First, we determined that rhodamine-labeled MCH (R-MCH), when microinjected into the lateral ventricle, is internalized in serotonergic and non-serotonergic DRN neurons in rats and cats. These data strongly suggest that these neurons express MCHergic receptors. Second, in rats, we demonstrated that the microinjection of MCH into the lateral ventricle results in a significant decrease in the firing rate in 59% of the neurons recorded in the DRN; the juxtacellular administration of MCH reduced the discharge in 80% of these neurons. Some of the neurons affected by MCH were likely serotonergic on the basis of their electrophysiological and pharmacological properties. We conclude that MCH reduces the activity of serotonergic neurons of the DRN. These and previous data suggest that the MCHergic modulation of serotonergic activity within the DRN is involved in the regulation of REM sleep as well as in the pathophysiology of depressive disorders.


Subject(s)
Dorsal Raphe Nucleus/drug effects , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Neurons/drug effects , Pituitary Hormones/administration & dosage , Action Potentials/drug effects , Animals , Cats , Dorsal Raphe Nucleus/physiology , Glutamate Decarboxylase/metabolism , Immunohistochemistry , Microelectrodes , Microinjections , Neurons/physiology , Photomicrography , Rats, Wistar , Rhodamines
17.
Brain Res Bull ; 107: 61-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25048146

ABSTRACT

Some members of the RFamide peptide family are associated with feeding in rodents. For example, neuropeptide FF and prolactin-releasing peptide cause anorexigenic, while 26RFa and QRFP result in orexigenic effects. I.c.v. microinjection of RFamide-related peptide-3 (RFRP-3) facilitates feeding. Feeding related effects of RFRP-3, however, have not been studied after direct brain microinjections in rats. The central part of amygdala (CeA) is essentially involved in the regulation of feeding and body weight. RFRP-3 positive nerve cells were detected in the rat hypothalamus and RFRP-3 immunoreactive fibers were identified in the CeA. RFRP analogs bind with relatively high affinity to the NPFF1 and NPFF2 receptors (NPFF-R). RFRP-3 has potent activity for NPFF-1 that is expressed in the CeA. To evaluate the role of RFRP-3 in feeding regulation rats were microinjected with different doses of RFRP-3 and their food intake were quantified over a 60 min period. Liquid food intake of male Wistar rats was measured after bilateral intraamygdalar administration of RFRP-3 (25, 50, 100 or 200 ng/side, RFRP-3 dissolved in 0.15M sterile NaCl/0.4 µl, respectively). The 50 ng and 100 ng doses of RFRP-3 microinjections resulted in significant decrease of food intake. Twenty-five and 200 ng had no effect. Food intake decreasing effect of RFRP-3 was eliminated by NPFF-R antagonist RF9 pretreatment. In open-field test effective doses of RFRP-3 did not modify spontaneous locomotor activity and general behavior of animals did not change. Our results are the first reporting that RFRP-3 injected to the CeA resulted in a decrease of liquid food consumption. This is a receptor-linked effect because it was eliminated by NPFF-R antagonist.


Subject(s)
Central Amygdaloid Nucleus/physiology , Eating/physiology , Hypothalamic Hormones/physiology , Animals , Central Amygdaloid Nucleus/drug effects , Eating/drug effects , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/pharmacology , Male , Microinjections , Motor Activity/drug effects , Rats , Rats, Wistar
18.
Gen Comp Endocrinol ; 205: 49-54, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24698787

ABSTRACT

The action of testosterone on male socio-sexual behaviors, such as aggressive and sexual behaviors, requires its aromatization into estrogen (neuroestrogen) in the brain. Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotropin secretion from the pituitary. On the other hand, wide distribution of GnIH-immunoreactive (ir) neuronal fibers in the brain suggested their roles in the regulation of behavior. Our recent studies have shown that GnIH indeed inhibits aggressive and sexual behaviors. Accordingly, we further investigated the effect of GnIH on aromatase activity and estrogen synthesis in the brain. Abundant GnIH-ir neuronal fibers were observed in the vicinity of aromatase-ir cells in the brain, such as in the preoptic area (POA) that is thought to be the most critical site of aromatization and neuroestrogen action for the regulation of socio-sexual behavior. GnIH receptor (GPR147) mRNA was also expressed in aromatase-ir cells in the POA. GnIH stimulated the activity of aromatase and increased neuroestrogen synthesis in the POA through GPR147. The increase in neuroestrogen concentration in the POA was associated with a significant decrease in aggressive behavior. Finally, centrally administered 17ß-estradiol at higher doses inhibited aggressive behavior. These findings indicated that GnIH inhibits aggressive behavior by directly activating aromatase and increasing neuroestrogen synthesis in the brain beyond its optimum concentration for the expression of aggressive behavior. This review highlights recent findings of the role of GnIH in the regulation of neuroestrogen synthesis and its possible function in the regulation of socio-sexual behaviors.


Subject(s)
Aggression/drug effects , Brain/drug effects , Brain/metabolism , Estrogens/biosynthesis , Hypothalamic Hormones/pharmacology , Neuropeptides/biosynthesis , Quail/metabolism , Animals , Hypothalamic Hormones/administration & dosage , Male
19.
Nat Commun ; 5: 3061, 2014.
Article in English | MEDLINE | ID: mdl-24430094

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotropin secretion and socio-sexual behaviours. Oestrogen (neuroestrogen) synthesized in the brain from androgen by aromatase regulates male socio-sexual behaviours. Here we show that GnIH directly activates aromatase and increases neuroestrogen synthesis in the preoptic area (POA) and inhibits socio-sexual behaviours of male quail. Aromatase activity and neuroestrogen concentration in the POA are low in the morning when the birds are active, but neuroestrogen synthesis gradually increases until the evening when the birds become inactive. Centrally administered GnIH in the morning increases neuroestrogen synthesis in the POA and decreases socio-sexual behaviours. Centrally administered 17ß-oestradiol at higher doses also inhibits socio-sexual behaviours in the morning. These results suggest that GnIH inhibits male socio-sexual behaviours by increasing neuroestrogen synthesis beyond its optimum concentration for the expression of socio-sexual behaviours. This is the first demonstration of any hypothalamic neuropeptide that directly regulates neuroestrogen synthesis.


Subject(s)
Behavior, Animal/physiology , Coturnix/physiology , Estrogens/metabolism , Hypothalamus/physiology , Preoptic Area/metabolism , Sexual Behavior, Animal/physiology , Androgens/metabolism , Animals , Aromatase/metabolism , Behavior, Animal/drug effects , Circadian Rhythm/physiology , Dose-Response Relationship, Drug , Estradiol/pharmacology , Glycoproteins/administration & dosage , Glycoproteins/pharmacology , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/pharmacology , Infusions, Intraventricular , Male , Models, Animal , Sexual Behavior, Animal/drug effects
20.
Peptides ; 50: 96-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24126282

ABSTRACT

An emerging body of evidence involves the hypothalamic neuropeptide melanin-concentrating hormone (MCH) in the regulation of emotional states. We have reported a pro-depressive effect induced by MCH after its microinjection into the dorsal raphe nucleus (DR) evaluated in the forced swimming test (FST) in rats. Here we extended this study to the median raphe nucleus (MnR). Firstly, the presence of MCH-containing fibers in the rat MnR was analyzed by means of immunohistochemistry. Secondly, the behavioral effect induced by the microinjection of MCH into the MnR was assessed using the FST. Morphological results showed a large density of MCHergic fibers within the MnR. Behavioral results indicated that 100 ng of MCH (but not 50 ng) significantly increased the immobility time and decreased the swimming time, demonstrating a depressive-like effect. In contrast, climbing behavior was not significantly affected. Present findings revealed that the MnR neurons participate in the MCHergic control of affective-related behavioral responses. However, the behavioral patterns induced by MCH in the MnR and DR were different. This could be explained by anatomical and physiological differences between both nuclei.


Subject(s)
Behavior, Animal/drug effects , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Pituitary Hormones/administration & dosage , Raphe Nuclei/drug effects , Animals , Depression, Chemical , Immunohistochemistry , Male , Microinjections , Organ Specificity , Raphe Nuclei/anatomy & histology , Raphe Nuclei/physiology , Rats , Rats, Wistar , Swimming/psychology
SELECTION OF CITATIONS
SEARCH DETAIL