Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Peptides ; 126: 170249, 2020 04.
Article in English | MEDLINE | ID: mdl-31911169

ABSTRACT

Serotonergic neurons of the median raphe nucleus (MnR) and hypothalamic melanin-concentrating hormone (MCH)-containing neurons, have been involved in the control of REM sleep and mood. In the present study, we examined in rats and cats the anatomical relationship between MCH-containing fibers and MnR neurons, as well as the presence of MCHergic receptors in these neurons. In addition, by means of in vivo unit recording in urethane anesthetized rats, we determined the effects of MCH in MnR neuronal firing. Our results showed that MCH-containing fibers were present in the central and paracentral regions of the MnR. MCHergic fibers were in close apposition to serotonergic and non-serotonergic neurons. By means of an indirect approach, we also analyzed the presence of MCHergic receptors within the MnR. Accordingly, we microinjected MCH conjugated with the fluorophore rhodamine (R-MCH) into the lateral ventricle. R-MCH was internalized into serotonergic and non-serotonergic MnR neurons; some of these neurons were GABAergic. Furthermore, we determined that intracerebroventricular administration of MCH induced a significant decrease in the firing rate of 53 % of MnR neurons, while the juxtacellular administration of MCH reduced the frequency of discharge in 67 % of these neurons. Finally, the juxtacellular administration of the MCH-receptor antagonist ATC-0175 produced an increase in the firing rate in 78 % of MnR neurons. Hence, MCH produces a strong regulation of MnR neuronal activity. We hypothesize that MCHergic modulation of the MnR neuronal activity may be involved in the promotion of REM sleep and in the pathophysiology of depressive disorders.


Subject(s)
Hypothalamic Hormones/pharmacology , Hypothalamus/drug effects , Melanins/pharmacology , Nerve Fibers/drug effects , Neurons/drug effects , Pituitary Hormones/pharmacology , Raphe Nuclei/drug effects , Receptors, Pituitary Hormone/metabolism , Animals , Cats , Hypothalamus/metabolism , Hypothalamus/physiology , Nerve Fibers/metabolism , Nerve Fibers/physiology , Neurons/metabolism , Neurons/physiology , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Rats , Rats, Wistar
2.
Nat Commun ; 10(1): 2505, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31175285

ABSTRACT

Brain signals that govern memory formation remain incompletely identified. The hypothalamus is implicated in memory disorders, but how its rapidly changing activity shapes memorization is unknown. During encounters with objects, hypothalamic melanin-concentrating hormone (MCH) neurons emit brief signals that reflect object novelty. Here we show that targeted optogenetic silencing of these signals, performed selectively during the initial object encounters (i.e. memory acquisition), prevents future recognition of the objects. We identify an upstream inhibitory microcircuit from hypothalamic GAD65 neurons to MCH neurons, which constrains the memory-promoting MCH cell bursts. Finally, we demonstrate that silencing the GAD65 cells during object memory acquisition improves future object recognition through MCH-receptor-dependent pathways. These results provide causal evidence that object-associated signals in genetically distinct but interconnected hypothalamic neurons differentially control whether the brain forms object memories. This gating of memory formation by hypothalamic activity establishes appropriate behavioral responses to novel and familiar objects.


Subject(s)
Glutamate Decarboxylase/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/physiology , Melanins/metabolism , Memory/physiology , Neurons/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/metabolism , Recognition, Psychology/physiology , Animals , Hypothalamus/cytology , Hypothalamus/metabolism , Memory/drug effects , Mice , Neural Inhibition/physiology , Neural Pathways , Optogenetics , Piperidines/pharmacology , Receptors, Pituitary Hormone/antagonists & inhibitors , Recognition, Psychology/drug effects
3.
Gene ; 615: 57-67, 2017 Jun 05.
Article in English | MEDLINE | ID: mdl-28300612

ABSTRACT

Melanin-concentrating hormone (MCH) is a neuropeptide expressed in the brain and exerts its actions through interaction with the two known G protein-coupled receptors, namely melanin-concentrating hormone receptor 1 and 2 (MCHR1 and MCHR2) in mammals. However, the information regarding the expression and functionality of MCH and MCHR(s) remains largely unknown in birds. In this study, using RT-PCR and RACE PCR, we amplified and cloned a MCHR1-like receptor, which is named cMCHR4 according to its evolutionary origin, and a MCHR2 from chicken brain. The cloned cMCHR4 was predicted to encode a receptor of 367 amino acids, which shares high amino acid identities with MCHR4 of ducks (90%), western painted turtles (85%), and coelacanths (77%), and a comparatively low identity to human MCHR1 (58%) and MCHR2 (38%), whereas chicken MCHR2 encodes a putative C-terminally truncated receptor and is likely a pseudogene. Using cell-based luciferase reporter assays or Western blot, we further demonstrated that chicken (and duck) MCHR4 could be potently activated by chicken MCH1-19, and its activation can elevate calcium concentration and activate MAPK/ERK and cAMP/PKA signaling pathways, indicating an important role of MCHR4 in mediating MCH actions in birds. Quantitative real-time PCR revealed that both cMCH and cMCHR4 mRNA are expressed in various brain regions including the hypothalamus, and cMCH expression in the hypothalamus of 3-week-old chicks could be induced by 36-h fasting, indicating that cMCH expression is correlated with energy balance. Taken together, characterization of chicken MCH and MCHR4 will aid to uncover the conserved roles of MCH across vertebrates.


Subject(s)
Chickens/genetics , Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Melanins/genetics , Pituitary Hormones/genetics , Receptors, Pituitary Hormone/genetics , Animals , Cloning, Molecular , Ducks/genetics , Fasting , Gene Expression Regulation , HEK293 Cells , Humans , Hypothalamic Hormones/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/metabolism , Up-Regulation
4.
Neuropeptides ; 64: 123-130, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27614713

ABSTRACT

Hypothalamic alpha-melanocyte-stimulating hormone (α-MSH) is a key catabolic mediator of energy homeostasis. Its anorexigenic and hypermetabolic effects show characteristic age-related alterations that may be part of the mechanism of middle-aged obesity and geriatric anorexia/cachexia seen in humans and other mammals. We aimed to investigate the role of α-MSH in mitochondrial energy metabolism during the course of aging in a rodent model. To determine the role of α-MSH in mitochondrial energy metabolism in muscle, we administered intracerebroventricular (ICV) infusions of α-MSH for 7-days to different age-groups of male Wistar rats. The activities of oxidative phosphorylation complexes I to V and citrate synthase were determined and compared to those of age-matched controls. We also quantified mitochondrial DNA (mtDNA) copy number and measured the expression of the master regulators of mitochondrial biogenesis, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and peroxisome proliferator-activated receptor gamma (PPARγ). The peptide reduced weight gain in juvenile rats to one fifth of that of controls and increased the weight loss in older animals by about five fold. Mitochondrial DNA copy number inversely correlated with changes in body weight in controls, but not in α-MSH-treated animals. The strong increase in body weight in young rats was associated with a low mtDNA copy number and high PPARγ mRNA levels in controls. Expression of PGC-1α and PPARγ declined with age, whereas OXPHOS and citrate synthase enzyme activities were unchanged. In contrast, α-MSH treatment suppressed OXPHOS enzyme and citrate synthase activity. In conclusion, our results showed age-related differences in the metabolic effects of α-MSH. In addition, administration of α-MSH suppressed citrate synthase and OXPHOS activities independent of age. These findings suggest that α-MSH exposure may inhibit mitochondrial biogenesis.


Subject(s)
Energy Metabolism/drug effects , Mitochondria/drug effects , Muscle, Skeletal/metabolism , alpha-MSH/metabolism , Aging , Animals , Hypothalamus/metabolism , Male , PPAR gamma/metabolism , Rats, Wistar , Receptors, Pituitary Hormone/drug effects , Receptors, Pituitary Hormone/metabolism , Transcription Factors/metabolism
5.
J Neuroendocrinol ; 25(6): 570-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23414264

ABSTRACT

Previous studies have shown that oestradiol (E2) decreases the orexigenic effect of melanin-concentrating hormone (MCH). In the present study, we examined whether this action of E2 is mediated by its ability to decrease the expression of MCH or its receptor (MCHR1). Using immunocytochemistry and western blotting, we examined whether E2 decreases MCH-immunoreactive neurones or MCHR1 protein content in the hypothalamus of female rats. We found that both MCH and MCHR1 protein expression was decreased by acute E2 treatment in ovariectomised rats, and by the peri-ovulatory increase in circulating E2 in pro-oestrous rats, relative to rats at other cycle stages. To determine whether these changes in MCH/MCHR1 protein expression may be mediated by E2's ability to directly regulate the transcription of MCH and MCHR1 genes, the effect of E2 treatment on MCH and MCHR1 mRNA expression in a neuronal hypothalamic cell line was examined using real-time reverse transcriptase-polymerase chain reaction. We also determined whether MCH and oestrogen receptor (ER)α are co-expressed in the hypothalamus of female rats. E2 treatment did not decrease MCH or MCHR1 mRNA expression in vitro, and no hypothalamic neurones were identified that co-expressed MCH and ERα. We conclude that E2-dependent decreases in hypothalamic MCH/MCHR1 protein expression mediate the ability of E2 to decrease MCH-induced feeding. The current findings suggest, however, that E2 exerts these actions indirectly, most likely though interactions with other neuronal systems that provide afferent input to MCH and MCHR1 neurones.


Subject(s)
Estradiol/pharmacology , Hypothalamic Hormones/metabolism , Hypothalamus/drug effects , Melanins/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/metabolism , Animals , Base Sequence , DNA Primers , Estrogen Receptor alpha/physiology , Estrus , Female , Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Melanins/genetics , Pituitary Hormones/genetics , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction , Receptors, Pituitary Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Gen Comp Endocrinol ; 179(1): 78-87, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22884735

ABSTRACT

Melanin-concentrating hormone (MCH) is a neuromodulator, synthesized in the hypothalamus, that regulates both appetite and energy homeostasis in mammals. MCH was initially identified in teleost fishes as a pituitary gland hormone that induced melanin aggregation in chromatophores in the skin; however, this function of MCH has not been observed in other vertebrates. Recent studies suggest that MCH is involved in teleost feeding behavior, spurring the hypothesis that the original function of MCH in early vertebrates was appetite regulation. The present study reports the results of cDNAs cloning encoding preproMCH and two MCH receptors from an elasmobranch fish, Sphyrna lewini, a member of Chondrichthyes, the earliest diverged class in gnathostomes. The putative MCH peptide is composed of 19 amino acids, similar in length to the mammalian MCH. Reverse-transcription polymerase chain reaction revealed that MCH is expressed in the hypothalamus in S. lewini MCH cell bodies and fibers were identified by immunochemistry in the hypothalamus, but not in the pituitary gland, suggesting that MCH is not released via the pituitary gland into general circulation. MCH receptor genes mch-r1 and mch-r2 were expressed in the S. lewini hypothalamus, but were not found in the skin. These results indicate that MCH does not have a peripheral function, such as a melanin-concentrating effect, in the skin of S. lewini hypothalamic MCH mRNA levels were not affected by fasting, suggesting that feeding conditions might not affect the expression of MCH in the hypothalamus.


Subject(s)
Fish Proteins/chemistry , Hypothalamic Hormones/chemistry , Melanins/chemistry , Pituitary Hormones/chemistry , Receptors, Pituitary Hormone/chemistry , Sharks/genetics , Amino Acid Sequence , Animals , Brain/metabolism , Cloning, Molecular , DNA, Complementary/chemistry , Fish Proteins/genetics , Fish Proteins/metabolism , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/genetics , Melanins/metabolism , Molecular Sequence Data , Phylogeny , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , RNA, Messenger/chemistry , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Sequence Alignment , Sequence Analysis, Protein , Sharks/metabolism , Skin/metabolism
7.
Bioorg Med Chem Lett ; 22(11): 3781-5, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22542010

ABSTRACT

An initial SAR study resulted in the identification of the novel, potent MCHR1 antagonist 2. After further profiling, compound 2 was discovered to be a potent inhibitor of the hERG potassium channel, which prevented its further development. Additional optimization of this structure resulted in the discovery of the potent MCHR1 antagonist 11 with a dramatically reduced hERG liability. The decrease in hERG activity was confirmed by several in vivo preclinical cardiovascular studies examining QT prolongation. This compound demonstrated good selectivity for MCHR1 and possessed good pharmacokinetic properties across preclinical species. Compound 11 was also efficacious in reducing body weight in two in vivo mouse models. This compound was selected for clinical evaluation and was given the code AMG 076.


Subject(s)
Carbazoles/chemistry , Cyclohexanecarboxylic Acids/chemistry , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Receptors, Pituitary Hormone/antagonists & inhibitors , Animals , Body Weight/drug effects , Brain/drug effects , Brain/metabolism , Carbazoles/chemical synthesis , Carbazoles/pharmacokinetics , Cyclohexanecarboxylic Acids/chemical synthesis , Cyclohexanecarboxylic Acids/pharmacokinetics , Diet, High-Fat , Dogs , Drug Evaluation, Preclinical , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Structure-Activity Relationship
8.
Mol Cell Endocrinol ; 348(1): 281-96, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-21945816

ABSTRACT

In order to better understand the role of melanin-concentrating hormone (MCH) in the regulation of appetite in fish, the mRNAs of two forms of MCH, prepro-MCH and MCH2, and two forms of MCH receptors, MCH-R1 and MCH-R2, were isolated from winter flounder (Pseudopleuronectes americanus). In addition, the mRNA expressions of these peptides and their receptors were determined under fed and fasted conditions. Both MCHs are expressed in forebrain and midbrain, as well as peripheral tissues including gut and gonads. Both MCH-Rs are ubiquitously expressed in the brain and periphery. Fasting induced an increase in the expression levels of MCH and MCH-R1 mRNAs in optic tectum/thalamus and hypothalamus but had no effect on either MCH2 or MCH-R2 mRNA expressions. Our results suggest that MCH and MCH-R1, but not MCH2 and MCH-R2 might have a role in the regulation of appetite in flounder.


Subject(s)
Appetite Regulation , Fish Proteins/physiology , Flounder/physiology , Hypothalamic Hormones/physiology , Melanins/physiology , Pituitary Hormones/physiology , Receptors, Pituitary Hormone/physiology , Amino Acid Sequence , Animals , Brain/metabolism , Female , Fish Proteins/genetics , Fish Proteins/metabolism , Flounder/genetics , Flounder/metabolism , Food Deprivation , Gene Expression , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Male , Melanins/genetics , Melanins/metabolism , Molecular Sequence Data , Organ Specificity , Phylogeny , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid
9.
Phytother Res ; 24(6): 919-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20013814

ABSTRACT

The time-resolved fluorescence technique based on melanin-concentrating hormone (MCH) receptor subtype-1 (MCH-1 receptor) binding assay was adopted to carry out a bioassay-guided fractionation of the methanol extract of Morus alba leaves. This fractionation and purification led to the isolation of two compounds identified as pheophorbide a methyl ester and 13(2)(S)-hydroxypheophorbide a methyl ester. These active pheophorbides exhibited potent inhibitory activity in binding of europium-labeled MCH to the human recombinant MCH-1 receptor (IC(50) value; 4.03 and 0.33 microM, respectively). Besides binding activity, the pheophorbides inhibited MCH-mediated extracellular signal-regulated kinase (ERK) phosphorylation in Chinese hamster ovary cells expressing human MCH-1 receptor. These results suggest that pheophorbide a methyl ester and 13(2)(S)-hydroxypheophorbide a methyl ester act as modulators of MCH-1 receptor and MCH-mediated ERK signaling.


Subject(s)
Chlorophyll/analogs & derivatives , Morus/chemistry , Plant Extracts/pharmacology , Receptors, Pituitary Hormone/metabolism , Animals , CHO Cells , Chlorophyll/pharmacology , Cricetinae , Cricetulus , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Molecular Structure , Phosphorylation , Plant Leaves/chemistry , Recombinant Proteins/metabolism , Signal Transduction
10.
J Comp Neurol ; 517(5): 695-710, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19827161

ABSTRACT

Melanin-concentrating hormone (MCH) regulates feeding and complex behaviors in mammals and pigmentation in fish. The relationship between fish and mammalian MCH systems is not well understood. Here, we identify and characterize two MCH genes in zebrafish, Pmch1 and Pmch2. Whereas Pmch1 and its corresponding MCH1 peptide resemble MCH found in other fish, the zebrafish Pmch2 gene and MCH2 peptide share genomic structure, synteny, and high peptide sequence homology with mammalian MCH. Zebrafish Pmch genes are expressed in closely associated but non-overlapping neurons within the hypothalamus, and MCH2 neurons send numerous projections to multiple MCH receptor-rich targets with presumed roles in sensory perception, learning and memory, arousal, and homeostatic regulation. Preliminary functional analysis showed that whereas changes in zebrafish Pmch1 expression correlate with pigmentation changes, the number of MCH2-expressing neurons increases in response to chronic food deprivation. These findings demonstrate that zebrafish MCH2 is the putative structural and functional ortholog of mammalian MCH and help elucidate the nature of MCH evolution among vertebrates.


Subject(s)
Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Melanins/genetics , Neurons/metabolism , Pituitary Hormones/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Base Sequence , Gene Expression Regulation , Hypothalamic Hormones/metabolism , Hypothalamus/cytology , In Situ Hybridization , Melanins/metabolism , Molecular Sequence Data , Neurons/cytology , Pigmentation/genetics , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Sequence Homology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
11.
Results Probl Cell Differ ; 46: 159-79, 2008.
Article in English | MEDLINE | ID: mdl-18227983

ABSTRACT

Melanin-concentrating hormone (MCH) is a neuropeptide that was originally isolated from salmon pituitary where it causes pigment aggregation. MCH is also abundantly present in mammalian neurons and expressed in the lateral hypothalamus and zona incerta, brain regions that are known to be at the center of feeding behavior. MCH binds to and activates two G protein-coupled receptors, MCH1R and MCH2R. Although MCH2R is non-functional in rodents, genetic and pharmacological studies have demonstrated that rodent MCH1R is involved in the regulation of feeding behavior and energy balance. Unexpectedly, some antagonists have provided evidence that MCH signaling participates in the regulation of other processes, such as emotion and stress. The discovery of MCH receptors has extensively promoted the progress of MCH studies and may represent an ideal example of how deorphanized receptors can open new directions toward more detailed physiological studies.


Subject(s)
Hypothalamic Hormones/physiology , Melanins/physiology , Pituitary Hormones/physiology , Amino Acid Sequence , Animals , Humans , Hypothalamus/metabolism , Mice , Models, Biological , Molecular Sequence Data , Pituitary Gland/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Somatostatin/metabolism , Salmon/metabolism , Sequence Homology, Amino Acid , Signal Transduction
12.
Curr Top Med Chem ; 7(15): 1440-54, 2007.
Article in English | MEDLINE | ID: mdl-17897031

ABSTRACT

Melanin concentrating hormone (MCH) is a cyclic, nonadecapeptide expressed in the CNS of all vertebrates that regulates feeding behavior and energy homeostasis. The MCH-1 receptor (MCH-R1) has been identified as a key target in MCH regulation, as small molecule antagonists of MCH-R1 have demonstrated activity in vivo. Herein, we chronicle our efforts to optimize a hit identified via high throughput screening of our proprietary compound library. Several challenges such as selectivity over other receptors, toxicity of a potential metabolite and determining receptor occupancy via a medium throughput assay will be reviewed.


Subject(s)
Obesity/drug therapy , Obesity/metabolism , Receptors, Pituitary Hormone/antagonists & inhibitors , Receptors, Pituitary Hormone/metabolism , Animals , Drug Evaluation, Preclinical , Humans , Obesity/pathology , Receptors, Pituitary Hormone/classification , Structure-Activity Relationship , Time Factors
13.
J Biol Chem ; 281(43): 32496-507, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16945926

ABSTRACT

Screening of a human brain cDNA library using the C-terminal tail of the melanin-concentrating hormone receptor 1 (MCHR1) as bait in a yeast two-hybrid assay resulted in the identification of the neurite-outgrowth related factor, neurochondrin. This interaction was verified in overlay, pulldown, and co-immunoprecipitation assays. Deletion mapping confined the binding to the C terminus of neurochondrin and to the proximal C terminus of MCHR1, a region known to be involved in G protein binding and signal transduction. This region of the MCHR1 is also able to interact with the actin- and intermediate filament-binding protein, periplakin. Interactions of MCHR1 with neurochondrin and periplakin were competitive, indicating that these two proteins bind to overlapping regions of MCHR1. Although neurochondrin did not interfere with melanin-concentrating hormone-mediated internalization of the receptor, it did inhibit G protein-coupled signal transduction via both Galpha(i/o) and Galpha(q/11) family G proteins as measured by each of melanin-concentrating hormone-induced G protein-activated inwardly rectifying K(+) channel activity of voltage-clamped amphibian oocytes, by calcium mobilization in transfected mammalian cells, and by reduction in the capacity of melanin-concentrating hormone to promote binding of [(35)S]guanosine 5'-3-O-(thio)triphosphate to both Galpha(o1) and Galpha(11). Immunohistochemistry revealed co-expression of neurochondrin and MCHR1 within the rodent brain, suggesting that neurochondrin may be involved in the regulation of MCHR1 signaling and play a role in modulating melanin-concentrating hormone-mediated functions in vivo.


Subject(s)
GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Pituitary Hormone/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites , Biotinylation , COS Cells , Cell Line , Chlorocebus aethiops , DNA, Complementary/metabolism , Escherichia coli/genetics , Humans , Immunohistochemistry , Protein Binding , Rats , Receptors, Pituitary Hormone/chemistry , Receptors, Pituitary Hormone/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection
14.
Brain Res Mol Brain Res ; 128(2): 150-9, 2004 Sep 28.
Article in English | MEDLINE | ID: mdl-15363890

ABSTRACT

Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide that stimulates feeding and increases body weight in rodents. We studied the role of the system in energy homeostasis and its regulation by the satiety signals, leptin and insulin. We used real-time PCR to measure the hypothalamic expression of MCH and its receptor (MCHR1) in two contrasting models of altered nutritional status, namely, obesity induced by 8 weeks' voluntary overeating and food restriction for 10 days. Diet-fed rats were stratified according to final total fat-pad mass into a 'high fat gain' group (HG) and 'low fat gain' group (LG). MCH mRNA levels were increased by 31% (p>0.05) and 49% (p<0.05) in the LG and HG, respectively, compared with controls. MCHR1 mRNA levels rose by 118% in the LG (p<0.01) and 85% in the HG (p<0.01). There were significant positive correlations (p<0.05) between plasma leptin concentration and both MCH and MCHR1 mRNA levels, and between plasma insulin and MCHR1 expression. A positive correlation was also observed between MCH and MCHR1 mRNA levels (p<0.05). Food-restricted rats showed no significant alterations in the levels of either MCH mRNA or MCHR1 mRNA. In a second experiment, we measured MCH peptide levels in five discrete hypothalamic areas of dietary-obese rats. MCH concentrations were significantly increased in the arcuate nuclei of the HG (p<0.05) and the paraventricular nuclei of both the LG (p<0.05) and HG (p<0.05), compared with their lean counterparts. These results suggest that the MCH system becomes more active in dietary obesity and could be involved in enhancing appetite for palatable food. The possibility that MCH and MCHR1 expression are positively regulated by leptin and insulin, which normally inhibit feeding, is a putative explanation for how appetite for palatable food is able to override mechanisms that prevent the development of obesity.


Subject(s)
Diet/adverse effects , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Obesity/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/metabolism , Animals , Blood Glucose , Body Mass Index , Body Weight , Hypothalamic Hormones/genetics , Insulin/blood , Leptin/blood , Male , Melanins/genetics , Obesity/chemically induced , Pituitary Hormones/genetics , RNA, Messenger/biosynthesis , Radioimmunoassay/methods , Rats , Receptors, Pituitary Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
15.
Phytomedicine ; 11(7-8): 633-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15636177

ABSTRACT

The fixed valerian-hops extract combination Ze91019 is used as a sleep aid. Although its exact mechanism of action is not well understood, earlier studies indicate that the CNS effect of valerian might occur through interaction with the GABA, melatonin and/or the adenosine systems in the brain. The use of hops in sleep remedies, however, is mainly based on traditional use and scarce scientific information. In this report, the binding of Ze91019, and the component valerian and hops extracts within, was tested on 14 subtypes of five classes of central receptors (dopamine, serotonin, melatonin, MCH and neuropeptide-Y). Binding affinities could be demonstrated at some of the screened melatonin (ML1 and ML2) and serotonin (5-HT4e, 5-HT6 and 5-HT7) receptor subtypes.


Subject(s)
Humulus/chemistry , Hypnotics and Sedatives/metabolism , Plant Extracts/metabolism , Receptors, Cell Surface/metabolism , Valerian/chemistry , Animals , Binding, Competitive , Chickens , Cricetinae , Drug Combinations , Humans , In Vitro Techniques , Logistic Models , Receptors, Dopamine/metabolism , Receptors, Melatonin/metabolism , Receptors, Neuropeptide Y/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Serotonin/metabolism
16.
Gen Comp Endocrinol ; 132(3): 485-98, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12849972

ABSTRACT

In most amphibians, [Arg(8)] vasotocin (VT) has an antidiuretic effect that is coupled to the activation of adenylate cyclase. In contrast, mesotocin (MT) has a diuretic effect and acts via the inositol phosphate/calcium signaling pathway in amphibians. To further clarify the mechanisms of VT and MT activation, we report the molecular cloning of a VT receptor (VTR) and a MT receptor (MTR) from the Japanese tree frog, Hyla japonica. Tree frog VTR or MTR cDNA encoded 363 or 389 amino acids, and their amino acid sequences revealed close similarity to the mammalian vasopressin V(2) (51-52% identity) or toad MT (94% identity) receptors, respectively. Using CHO-K1 cells transfected with tree frog VTR, we observed elevated concentrations of intracellular cAMP following exposure of the cells to VT or other neurohypophysial hormones, whereas the cells transfected with MTR did not exhibit altered cAMP concentrations. The cells transfected with VTR exhibited the following efficiency for cAMP accumulation: VT = hydrin 1 > or = vasopressin > or = hydrin 2 > MT = oxytocin > isotocin. VTR or MTR mRNA exhibits a single 2.2- or 5.5-kb transcription band, respectively, and both are expressed in various tissues. VTR mRNA is clearly expressed in brain, heart, kidney, pelvic patch of skin, and urinary bladder, whereas brain, fat body, heart, kidney, and urinary bladder express MTR mRNA. Specifically, VTR mRNA in the pelvic patch or MTR mRNA in the dorsal skin is present at elevated levels in the skin. Characteristic distribution of VTR and MTR on osmoregulating organs indicates the ligands for these receptors would mediate a variety of functions. Further, the distribution of VTR in the skin would make the regional difference on cutaneous water absorption in response to VT in the Japanese tree frog.


Subject(s)
Anura/genetics , Receptors, Pituitary Hormone/genetics , Receptors, Vasopressin/genetics , Vasotocin/metabolism , Amino Acid Sequence , Animals , Anura/metabolism , Base Sequence , Cloning, Molecular , Cyclic AMP/metabolism , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Molecular Sequence Data , Neuropeptides/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Vasopressin/metabolism , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology , Sequence Homology, Amino Acid , Tissue Distribution
17.
Diabetes ; 51(5): 1453-60, 2002 May.
Article in English | MEDLINE | ID: mdl-11978642

ABSTRACT

Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) activate two shared receptors, VPAC1 and VPAC2. Activation of VPAC1 has been implicated in elevating glucose output, whereas activation of VPAC2 may be involved in insulin secretion. A hypothesis that a VPAC2-selective agonist would enhance glucose disposal by stimulating insulin secretion without causing increased hepatic glucose production was tested using a novel selective agonist of VPAC2. This agonist, BAY 55-9837, was generated through site-directed mutagenesis based on sequence alignments of PACAP, VIP, and related analogs. The peptide bound to VPAC2 with a dissociation constant (K(d)) of 0.65 nmol/l and displayed >100-fold selectivity over VPAC1. BAY 55-9837 stimulated glucose-dependent insulin secretion in isolated rat and human pancreatic islets, increased insulin synthesis in purified rat islets, and caused a dose-dependent increase in plasma insulin levels in fasted rats, with a half-maximal stimulatory concentration of 3 pmol/kg. Continuous intravenous or subcutaneous infusion of the peptide reduced the glucose area under the curve following an intraperitoneal glucose tolerance test. The peptide had effects on intestinal water retention and mean arterial blood pressure in rats, but only at much higher doses. BAY 55-9837 may be a useful therapy for the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Peptide Fragments/pharmacology , Receptors, Vasoactive Intestinal Peptide/agonists , Vasoactive Intestinal Peptide/pharmacology , Amino Acid Sequence , Animals , Blood Pressure/drug effects , Cells, Cultured , Diarrhea/drug therapy , Diarrhea/metabolism , Glucose/pharmacology , Heart Rate/drug effects , Hormones/blood , Humans , Injections, Intravenous , Injections, Subcutaneous , Insulin Secretion , Islets of Langerhans/drug effects , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Vasoactive Intestinal Peptide/analogs & derivatives , Vasoactive Intestinal Peptide/chemistry , Vasoactive Intestinal Peptide/metabolism
18.
Neurosci Lett ; 323(3): 179-82, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11959414

ABSTRACT

This study has demonstrated that the short and long form of the pituitary adenylate cyclase-activating polypeptide (PACAP), i.e. PACAP(27) and PACAP(38), moderately but significantly, and in a concentration (0.5-5 microM)-dependent manner, stimulated inositol phosphates (IPs) accumulation in myo-[(3)H]inositol-prelabeled cerebral cortical and hypothalamal slices of chick and duck, and in slices of rat cerebral cortex; both peptides had no effect on IPs formation in rat hypothalamus. Vasoactive intestinal peptide (VIP; 0.5-5 microM) weakly enhanced IPs accumulation in chick hypothalamus, had no significant action in chick cerebral cortex (in fact there was a tendency to attenuate the IPs response in this tissue), and slightly, but significantly, inhibited the IPs accumulation in rat cerebral cortex. VIP showed no activity in rat hypothalamus. It is concluded that the stimulatory action of PACAP on phosphoinositide metabolism in avian cerebral cortex, similar to rat cerebral cortex, is mediated via phospholipase C-linked PAC(1) type receptors. In chick hypothalamus, however, there may be a component of VPAC type receptors stimulating IPs formation.


Subject(s)
Birds/metabolism , Cerebral Cortex/enzymology , Hypothalamus/enzymology , Inositol Phosphates/metabolism , Neurons/enzymology , Neuropeptides/metabolism , Animals , Birds/anatomy & histology , Cerebral Cortex/cytology , Chickens , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Ducks , Hypothalamus/cytology , Male , Neurons/cytology , Neuropeptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/drug effects , Receptors, Pituitary Hormone/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
19.
Mol Pharmacol ; 60(4): 632-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562423

ABSTRACT

Using a genomics-based approach for screening orphan G-protein-coupled receptors, we have identified and cloned a novel high-affinity, melanin-concentrating hormone (MCH) receptor. This receptor, named S643b, displays the greatest overall identity (32%) with the previously reported human SLC-1 receptor (MCH1) and to a lesser extent with the somatostatin receptor subtypes. The gene encoding the S643b receptor spans more than 23 kilobase pairs (kb) and was mapped, by radiation hybrid experiments, on chromosome 6q14.3-q15. Comparison of the S643b cDNA with human genomic sequence reveals that the 340-amino-acid receptor is encoded by five exons. Its tissue distribution, as determined by Northern blot and reverse transcription-polymerase chain reaction analysis, indicates that a 4-kb transcript is predominantly expressed in the brain. When expressed in Chinese hamster ovary (CHO) cells, the S643b receptor displays a strong, dose-dependent, transient elevation of intracellular calcium in response to MCH (EC(50) = 9.5 nM). During the present study, we isolated a splice variant, designated S643a, encoding for a receptor that was not activated by MCH in a cellular calcium mobilization assay. Comparative pharmacological studies using CHO cells stably expressing either SLC-1 or S643b receptors demonstrated that similar structural features of MCH are required to stimulate intracellular Ca(2+) mobilization at both receptors. The identification and localization of this new MCH receptor (MCH2) provides further insight into the physiological implication of MCH in modulating behavioral responses, including food intake.


Subject(s)
Chromosomes, Human, Pair 6 , Receptors, Pituitary Hormone/genetics , Receptors, Somatostatin/metabolism , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/analysis , Humans , Molecular Sequence Data , Peptides/pharmacology , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled , Receptors, Pituitary Hormone/metabolism , Receptors, Somatostatin/chemistry , Sequence Homology, Amino Acid , Tissue Distribution
20.
J Mol Neurosci ; 14(3): 137-46, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10984189

ABSTRACT

To elucidate the functional role of the second extracellular loop of human vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating polypeptide (VIP/PACAP) receptor (hVPAC1R), surface expression, ligand binding, and receptor activation were analyzed. Amino acids in the entire second extracellular loop were individually substituted by alanine by site-directed mutagenesis. The mutant and wild-type receptors were transiently expressed in HEK293 cells and purified cell membranes were tested for the ability to bind VIP, while the receptor activity was measured as potency of cAMP production analysed on intact cells. Surface expression of the substituted conserved residues, W286A, I289A, W294A, and W295A, was evidently decreased to 20-30% compared to the wild-type expression. W286A also showed an significantly reduced potency of cAMP production. Substituted residues as F280A, E281A, and G284A showed a significant reduction in the potency of stimulated cAMP production amounting to 8-46-fold, compared to the wild-type with unaffected surface expression and VIP binding. These results indicate that some residues in the second extracellular loop of the human VPAC1R participate in the active mechanism of a ligand-mediated response without being directly involved in the binding of VIP.


Subject(s)
Receptors, Pituitary Hormone/chemistry , Receptors, Pituitary Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide/metabolism , Binding Sites/physiology , Cells, Cultured , Cyclic AMP/metabolism , DNA, Complementary , Humans , Iodine Radioisotopes , Kidney/cytology , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I , Sequence Homology, Amino Acid , Transfection , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL