Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 364
Filter
1.
J Neuroendocrinol ; 35(11): e13354, 2023 11.
Article in English | MEDLINE | ID: mdl-37946684

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) and the homologous peptide, vasoactive intestinal peptide (VIP), participate in glucose homeostasis using insulinotropic and counterregulatory processes. The role of VIP receptor 2 (VPAC2R) in these opposing actions needs further characterization. In this study, we examined the participation of VPAC2R on basal glycemia, fasted levels of glucoregulatory hormones and on glycemia responses during metabolic and psychogenic stress using gene-deleted (Vipr2-/- ) female mice. The mean basal glycemia was significantly greater in Vipr2-/- in the fed state and after an 8-h overnight fast as compared to wild-type (WT) mice. Insulin tolerance testing following a 5-h fast (morning fast, 0.38 U/kg insulin) indicated no effect of genotype. However, during a more intense metabolic challenge (8 h, ON fast, 0.25 U/kg insulin), Vipr2-/- females displayed significantly impaired insulin hypoglycemia. During immobilization stress, the hyperglycemic response and plasma epinephrine levels were significantly elevated above basal in Vipr2-/- , but not WT mice, in spite of similar stress levels of plasma corticosterone. Together, these results implicate participation of VPAC2R in upregulated counterregulatory processes influenced by enhanced sympathoexcitation. Moreover, the suppression of plasma GLP-1 levels in Vipr2-/- mice may have removed the inhibition on hepatic glucose production and the promotion of glucose disposal by GLP-1. qPCR analysis indicated deregulation of central gene markers of PACAP/VIP signaling in Vipr2-/- , upregulated medulla tyrosine hydroxylase (Th) and downregulated hypothalamic Vip transcripts. These results demonstrate a physiological role for VPAC2R in glucose metabolism, especially during insulin challenge and psychogenic stress, likely involving the participation of sympathoadrenal activity and/or metabolic hormones.


Subject(s)
Receptors, Pituitary Hormone , Receptors, Vasoactive Intestinal Peptide , Mice , Female , Animals , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Gene Deletion , Vasoactive Intestinal Peptide/metabolism , Insulin/metabolism , Glucose , Glucagon-Like Peptide 1 , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/genetics
2.
J Mol Neurosci ; 73(9-10): 724-737, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37646964

ABSTRACT

Previous evidence shows that rapid changes occur in the brain following spinal cord injury (SCI). Here, we interrogated the expression of the neuropeptides pituitary adenylyl cyclase-activating peptide (PACAP), vasoactive intestinal peptides (VIP), and their binding receptors in the rat brain 24 h following SCI. Female Sprague-Dawley rats underwent thoracic laminectomy; half of the rats received a mild contusion injury at the level of the T10 vertebrate (SCI group); the other half underwent sham surgery (sham group). Twenty-four hours post-surgery, the hypothalamus, thalamus, amygdala, hippocampus (dorsal and ventral), prefrontal cortex, and periaqueductal gray were collected. PACAP, VIP, PAC1, VPAC1, and VPAC2 mRNA and protein levels were measured by real-time quantitative polymerase chain reaction and Western blot. In SCI rats, PACAP expression was increased in the hypothalamus (104-141% vs sham) and amygdala (138-350%), but downregulated in the thalamus (35-95%) and periaqueductal gray (58-68%). VIP expression was increased only in the thalamus (175-385%), with a reduction in the amygdala (51-68%), hippocampus (40-75%), and periaqueductal gray (74-76%). The expression of the PAC1 receptor was the least disturbed by SCI, with decrease expression in the ventral hippocampus (63-68%) only. The expression levels of VPAC1 and VPAC2 receptors were globally reduced, with more prominent reductions of VPAC1 vs VPAC2 in the amygdala (21-70%) and ventral hippocampus (72-75%). In addition, VPAC1 downregulation also extended to the dorsal hippocampus (69-70%). These findings demonstrate that as early as 24 h post-SCI, there are region-specific disruptions of PACAP, VIP, and related receptor transcript and protein levels in supraspinal regions controlling higher cognitive functions.


Subject(s)
Receptors, Pituitary Hormone , Spinal Cord Injuries , Female , Rats , Animals , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Rats, Sprague-Dawley , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Spinal Cord Injuries/metabolism , Brain/metabolism
3.
Article in English | MEDLINE | ID: mdl-32849267

ABSTRACT

Lactation is a complex physiological process, depending on orchestrated central and peripheral events, including substantial brain plasticity. Among these events is a novel expression of pro-melanin-concentrating hormone (Pmch) mRNA in the rodent hypothalamus, such as the ventral part of the medial preoptic area (vmMPOA). This expression reaches its highest levels around postpartum day 19 (PPD19), when dams transition from lactation to the weaning period. The appearance of this lactation-related Pmch expression occurs simultaneously with the presence of one of the Pmch products, melanin-concentrating hormone (MCH), in the serum. Given the relevance of the MPOA to maternal physiology and the contemporaneity between Pmch expression in this structure and the weaning period, we hypothesized that MCH has a role in the termination of lactation, acting as a mediator between central and peripheral changes. To test this, we investigated the presence of the MCH receptor 1 (MCHR1) and its gene expression in the mammary gland of female rats in different stages of the reproductive cycle. To that end, in situ hybridization, RT-PCR, RT-qPCR, nucleotide sequencing, immunohistochemistry, and Western blotting were employed. Although Mchr1 expression was detected in the epidermis and dermis of both diestrus and lactating rats, parenchymal expression was exclusively found in the functional mammary gland of lactating rats. The expression of Mchr1 mRNA oscillated through the lactation period and reached its maximum in PPD19 dams. Presence of MCHR1 was confirmed with immunohistochemistry with preferential location of MCHR1 immunoreactive cells in the alveolar secretory cells. As was the case for gene expression, the MCHR1 protein levels were significantly higher in PPD19 than in other groups. Our data demonstrate the presence of an anatomical basis for the participation of MCH peptidergic system on the control of lactation through the mammary gland, suggesting that MCH could modulate a prolactation action in early postpartum days and the opposite role at the end of the lactation.


Subject(s)
Lactation , Mammary Glands, Animal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Animals , Female , Immunohistochemistry , Male , Mammary Glands, Animal/growth & development , Rats , Rats, Long-Evans
4.
Brain Res ; 1719: 71-76, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31121161

ABSTRACT

Melanin concentrating hormone (MCH) is involved in the initiation of maternal behavior during the postpartum period. Virgin females also display some aspects of maternal care independent of the hormonal and neurochemical changes associated with pregnancy and parturition. Maternal behavior in virgin females is triggered by pups-generated chemosensory signals. We therefore examined the role of MCH in maternal-related behaviors in virgin mice and whether it involves chemosensory mechanisms. We used mice with germline knock-out of MCH receptor (MCHR1 KO) and mice with conditional ablation of MCH neurons (MCH cKO) using Cre-inducible diphtheria toxin (iDTR) system. We report that germline deletion of MCHR1 and ablation of MCH neurons impair spontaneous maternal behavior that is induced upon pups' exposure. The latency and duration to retrieve pups by MCHR1 KO and MCH cKO mice are longer than their control littermate mice. In support of this finding, we found that in the three-chamber social test, both MCHR1 KO and MCH cKO mice display a lack of interest in interacting with pups. Strikingly, however, we found that while MCHR1 KO mice were unable to detect pups' chemosensory signals and displayed impairment in general olfactory discrimination, MCH cKO mice exhibited normal olfactory function. Our findings indicate that the lack of MCHR1 or of normal MCH levels causes defects in maternal behavior in non-sensitized virgin mice, and that disruption of the olfactory signaling might not count for these defects.


Subject(s)
Maternal Behavior/physiology , Receptors, Somatostatin/genetics , Smell/genetics , Animals , Behavior, Animal/physiology , Female , Germ-Line Mutation , Hypothalamic Hormones/genetics , Male , Melanins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Pituitary Hormones/genetics , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction/physiology , Smell/physiology
5.
Gen Comp Endocrinol ; 264: 138-150, 2018 08 01.
Article in English | MEDLINE | ID: mdl-28647318

ABSTRACT

To evaluate the association of the melanotropic peptides and their receptors for morphological color change, we investigated the effects of changes in background color, between white and black, on xanthophore density in the scales and expression levels of genes for hormonal peptides and corresponding receptors (MCH-R2, MC1R, and MC5R) in goldfish (Carassius auratus). The xanthophore density in both dorsal and ventral scales increased after transfer from a white to black background. However, xanthophore density in dorsal scales increased after transfer from a black to white background, and that of ventral scales decreased after transfer from a black to black background, which served as the control. In the white-reared fish, melanin-concentrating hormone (mch) mRNA content in the brain was higher than that in black-reared fish, whereas proopiomelanocortin a (pomc-a) mRNA content in the pituitary was lower than that in the black-reared fish. Agouti-signaling protein (asp) mRNA was detected in the ventral skin but not in the dorsal skin. No difference was observed in the asp mRNA content between fish reared in white or black background, suggesting that ASP might not be associated with background color adaptation. In situ hybridization revealed that both mc1r and mc5r were expressed in the xanthophores in scales. The mRNA content of mc1r in scales did not always follow the background color change, whereas those of mc5r decreased in the white background and increased in the black background, suggesting that mc5r might be a major factor reinforcing the function of MSH in morphological color changes. White backgrounds increased mch mRNA content in the brain, but decreased mch-r2 mRNA content in the scales. These altered expression levels of melanotropin receptors might affect reactivity to melanotropins through long-term adaptation to background color.


Subject(s)
Gene Expression Regulation , Goldfish/genetics , Melanocyte-Stimulating Hormones/genetics , Pigmentation/genetics , Receptors, Pituitary Hormone/genetics , Animal Scales/metabolism , Animals , Brain/metabolism , Color , Goldfish/metabolism , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , Melanins/genetics , Melanins/metabolism , Melanocyte-Stimulating Hormones/metabolism , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Pituitary Hormone/metabolism , Skin/metabolism
6.
Obes Res Clin Pract ; 12(2): 158-166, 2018.
Article in English | MEDLINE | ID: mdl-29066024

ABSTRACT

BACKGROUND: Prader Willi Syndrome (PWS) is a syndromic form of obesity caused by a chromosomal aberration on chromosome 15q11.2-q13. Patients with a comparable phenotype to PWS not carrying the 15q11.2-q13 defect are classified as Prader Willi like (PWL). In literature, PWL patients do frequently harbor deletions at 6q16, which led to the identification of the single-minded 1 (SIM1) gene as a possible cause for the presence of obesity in these patients. However, our previous work in a PWL cohort showed a rather limited involvement of SIM1 in the obesity phenotype. In this paper, we investigated the causal role of the melanin-concentrating hormone receptor 2 (MCHR2) gene in PWL patients, as most of the reported 6q16 deletions also encompass this gene and it is suggested to be active in the control of feeding behavior and energy metabolism. METHODS: Copy number variation analysis of the MCHR2 genomic region followed by mutation analysis of MCHR2 was performed in a PWL cohort. RESULTS: Genome-wide microarray analysis of 109 patients with PWL did not show any gene harboring deletions on chromosome 6q16. Mutation analysis in 92 patients with PWL demonstrated three MCHR2 variants: p.T47A (c.139A>G), p.A76A (c.228T>C) and c.*16A>G. We identified a significantly higher prevalence of the c.228T>C C allele in our PWL cohort compared to previously published results and controls of the ExAC Database. CONCLUSION: Overall, our results are in line with some previously performed studies suggesting that MCHR2 is not a major contributor to human obesity and the PWL phenotype.


Subject(s)
DNA Copy Number Variations/genetics , Obesity/genetics , Prader-Willi Syndrome/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Pituitary Hormone/genetics , Sequence Analysis, DNA , Adolescent , Basic Helix-Loop-Helix Transcription Factors , Child , DNA Mutational Analysis , Energy Metabolism/genetics , Feeding Behavior/physiology , Female , Genome-Wide Association Study , Humans , Male , Microarray Analysis , Obesity/etiology , Phenotype , Prader-Willi Syndrome/complications
7.
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
8.
Exp Dermatol ; 26(6): 536-541, 2017 06.
Article in English | MEDLINE | ID: mdl-27306922

ABSTRACT

Alopecia areata (AA) is a common hair loss disorder of autoimmune aetiology, which often results in pronounced psychological distress. Understanding of the pathophysiology of AA is increasing, due in part to recent genetic findings implicating common variants at several genetic loci. To date, no study has investigated the contribution of copy number variants (CNVs) to AA, a prominent class of genomic variants involved in other autoimmune disorders. Here, we report a genomewide- and a candidate gene-focused CNV analysis performed in a cohort of 585 patients with AA and 1340 controls of Central European origin. A nominally significant association with AA was found for CNVs in the following five chromosomal regions: 4q35.2, 6q16.3, 9p23, 16p12.1 and 20p12.1. The most promising finding was a 342.5-kb associated region in 6q16.3 (duplications in 4/585 patients; 0/1340 controls). The duplications spanned the genes MCHR2 and MCHR2-AS1, implicated in melanin-concentrating hormone (MCH) signalling. These genes have not been implicated in previous studies of AA pathogenesis. However, previous research has shown that MCHR2 affects the scale colour of barfin flounder fish via the induction of melanin aggregation. AA preferentially affects pigmented hairs, and the hair of patients with AA frequently shows a change in colour when it regrows following an acute episode of AA. This might indicate a relationship between AA, pigmentation and MCH signalling. In conclusion, the present results provide suggestive evidence for the involvement of duplications in MCHR2 in AA pathogenesis.


Subject(s)
Alopecia Areata/genetics , DNA Copy Number Variations , Genome-Wide Association Study , Receptors, G-Protein-Coupled/genetics , Receptors, Pituitary Hormone/genetics , Adult , Belgium , Chromosome Mapping , Cohort Studies , Europe , Female , Genotype , Germany , Humans , Hypothalamic Hormones/metabolism , Male , Melanins/metabolism , Netherlands , Pigmentation , Pituitary Hormones/metabolism , Polymorphism, Single Nucleotide , Signal Transduction
9.
Eur J Pharmacol ; 796: 45-53, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-27986627

ABSTRACT

Melanin-concentrating hormone (MCH), a cyclic neuropeptide expressed predominantly in the lateral hypothalamus, plays an important role in the control of feeding behavior and energy homeostasis. Mice lacking MCH or MCH1 receptor are resistant to diet-induced obesity (DIO) and MCH1 receptor antagonists show potent anti-obesity effects in preclinical studies, indicating that MCH1 receptor is a promising target for anti-obesity drugs. Moreover, recent studies have suggested the potential of MCH1 receptor antagonists for treatment of non-alcoholic fatty liver disease (NAFLD). In the present study, we show the anti-obesity and anti-hepatosteatosis effect of our novel MCH1 receptor antagonist, Compound A. Repeated oral administration of Compound A resulted in dose-dependent body weight reduction and had an anorectic effect in DIO mice. The body weight lowering effect of Compound A was more potent than that of pair-feeding. Compound A also reduced lipid content and the expression level of lipogenesis-, inflammation-, and fibrosis-related genes in the liver of DIO mice. Conversely, intracerebroventricular infusion of MCH caused induction of hepatic steatosis as well as increase in body weight in high-fat diet-fed wild type mice, but not MCH1 receptor knockout mice. The pair-feeding study revealed the MCH-MCH1 receptor system affects hepatic steatosis through a mechanism that is independent of body weight change. Metabolome analysis demonstrated that Compound A upregulated lipid metabolism-related molecules, such as acylcarnitines and cardiolipins, in the liver. These findings suggest that our novel MCH1 receptor antagonist, Compound A, exerts its beneficial therapeutic effect on NAFLD and obesity through a central MCH-MCH1 receptor pathway.


Subject(s)
Anti-Obesity Agents/pharmacology , Diet, High-Fat/adverse effects , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/chemically induced , Obesity/drug therapy , Receptors, Pituitary Hormone/antagonists & inhibitors , Animals , Anti-Obesity Agents/therapeutic use , Body Weight/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Knockout Techniques , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Rats , Receptors, Pituitary Hormone/deficiency , Receptors, Pituitary Hormone/genetics
10.
Sci Rep ; 6: 28969, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27373344

ABSTRACT

In many vertebrate species visible melanin-based pigmentation patterns correlate with high stress- and disease-resistance, but proximate mechanisms for this trait association remain enigmatic. Here we show that a missense mutation in a classical pigmentation gene, melanocyte stimulating hormone receptor (MC1R), is strongly associated with distinct differences in steroidogenic melanocortin 2 receptor (MC2R) mRNA expression between high- (HR) and low-responsive (LR) rainbow trout (Oncorhynchus mykiss). We also show experimentally that cortisol implants increase the expression of agouti signaling protein (ASIP) mRNA in skin, likely explaining the association between HR-traits and reduced skin melanin patterning. Molecular dynamics simulations predict that melanocortin 2 receptor accessory protein (MRAP), needed for MC2R function, binds differently to the two MC1R variants. Considering that mRNA for MC2R and the MC1R variants are present in head kidney cells, we hypothesized that MC2R activity is modulated in part by different binding affinities of the MC1R variants for MRAP. Experiments in mammalian cells confirmed that trout MRAP interacts with the two trout MC1R variants and MC2R, but failed to detect regulation of MC2R signaling, possibly due to high constitutive MC1R activity.


Subject(s)
Gene Expression Regulation , Oncorhynchus mykiss/physiology , Receptor Activity-Modifying Proteins/metabolism , Receptor, Melanocortin, Type 2/biosynthesis , Receptors, Pituitary Hormone/metabolism , Stress, Physiological , Animals , Gene Expression , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Protein Binding , RNA, Messenger/biosynthesis , Receptors, Pituitary Hormone/genetics
11.
Physiol Behav ; 163: 239-244, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27195455

ABSTRACT

The post-oral actions of glucose stimulate intake and condition flavor preferences in rodents. Hypothalamic melanin-concentrating hormone (MCH) neurons are implicated in sugar reward, and this study investigated their involvement in glucose preference conditioning in mice. In Exp. 1 MCH receptor 1 knockout (KO) and C57BL/6 wildtype (WT) mice learned to prefer 8% glucose over an initially more-preferred non-nutritive 0.1% sucralose+saccharin (S+S) solution. In contrast, the KO and WT mice preferred S+S to 8% fructose, which is consistent with this sugar's weak post-oral reinforcing action. In Exp. 2 KO and WT mice were trained to drink a flavored solution (CS+) paired with intragastric (IG) infusion of 16% glucose and a different flavored solution (CS-) paired with IG water. Both groups drank more CS+ than CS- in training and preferred the CS+ to CS- in a 2-bottle test. These results indicate that MCH receptor signaling is not required for flavor preferences conditioned by the post-oral actions of glucose. This contrasts with other findings implicating MCH signaling in other types of sugar reward processing.


Subject(s)
Conditioning, Classical/physiology , Food Preferences/physiology , Glucose/administration & dosage , Receptors, Pituitary Hormone/deficiency , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Pituitary Hormone/genetics , Saccharin/administration & dosage , Sweetening Agents , Taste/physiology
12.
Endocrinol Nutr ; 63(6): 274-84, 2016.
Article in English, Spanish | MEDLINE | ID: mdl-27091627

ABSTRACT

Pituitary adenomas are uncommon, difficult to diagnose tumors whose heterogeneity and low incidence complicate large-scale studies. The Molecular Registry of Pituitary Adenomas (REMAH) was promoted by the Andalusian Society of Endocrinology and Nutrition (SAEN) in 2008 as a cooperative clinical-basic multicenter strategy aimed at improving diagnosis and treatment of pituitary adenomas by combining clinical, pathological, and molecular information. In 2010, the Spanish Society of Endocrinology and Nutrition (SEEN) extended this project to national level and established 6 nodes with common protocols and methods for sample and clinical data collection, molecular analysis, and data recording in a common registry (www.remahnacional.com). The registry combines clinical data with molecular phenotyping of the resected pituitary adenoma using quantitative real-time PCR of expression of 26 genes: Pituitary hormones (GH-PRL-LH-FSH-PRL-ACTH-CGA), receptors (somatostatin, dopamine, GHRH, GnRH, CRH, arginine-vasopressin, ghrelin), other markers (Ki67, PTTG1), and control genes. Until 2015, molecular information has been collected from 704 adenomas, out of 1179 patients registered. This strategy allows for comparative and relational analysis between the molecular profile of the different types of adenoma and the clinical phenotype of patients, which may provide a better understanding of the condition and potentially help in treatment selection. The REMAH is therefore a unique multicenter, interdisciplinary network founded on a shared database that provides a far-reaching translational approach for management of pituitary adenomas, and paves the way for the conduct of combined clinical-basic innovative studies on large patient samples.


Subject(s)
Adenoma/epidemiology , Endocrinology/organization & administration , Pituitary Neoplasms/epidemiology , Precision Medicine/trends , Registries , Translational Research, Biomedical/trends , Adenoma/chemistry , Adenoma/genetics , Adolescent , Adult , Aged , Child , Databases, Factual , Endocrinology/trends , Female , Gene Expression Profiling , Genetic Association Studies , Humans , Male , Middle Aged , Molecular Biology , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Pituitary Hormones/analysis , Pituitary Hormones/genetics , Pituitary Neoplasms/chemistry , Pituitary Neoplasms/genetics , RNA, Neoplasm/genetics , Real-Time Polymerase Chain Reaction , Receptors, Pituitary Hormone/analysis , Receptors, Pituitary Hormone/genetics , Societies, Medical , Spain/epidemiology , Young Adult
13.
Nat Commun ; 6: 8842, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26548314

ABSTRACT

Understanding of the genetic regulation of puberty timing has come largely from studies of rare disorders and population-based studies in women. Here, we report the largest genomic analysis for puberty timing in 55,871 men, based on recalled age at voice breaking. Analysis across all genomic variants reveals strong genetic correlation (0.74, P=2.7 × 10(-70)) between male and female puberty timing. However, some loci show sex-divergent effects, including directionally opposite effects between sexes at the SIM1/MCHR2 locus (Pheterogeneity=1.6 × 10(-12)). We find five novel loci for puberty timing (P<5 × 10(-8)), in addition to nine signals in men that were previously reported in women. Newly implicated genes include two retinoic acid-related receptors, RORB and RXRA, and two genes reportedly disrupted in rare disorders of puberty, LEPR and KAL1. Finally, we identify genetic correlations that indicate shared aetiologies in both sexes between puberty timing and body mass index, fasting insulin levels, lipid levels, type 2 diabetes and cardiovascular disease.


Subject(s)
Cardiovascular Diseases/genetics , Diabetes Mellitus, Type 2/genetics , Polycystic Ovary Syndrome/genetics , Puberty/genetics , Adolescent , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Height/genetics , Body Mass Index , Bone Density/genetics , Child , Cholesterol, HDL/blood , Extracellular Matrix Proteins/genetics , Female , Genome-Wide Association Study , Humans , Insulin/blood , Lumbar Vertebrae/diagnostic imaging , Male , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 2/genetics , Radiography , Receptors, G-Protein-Coupled/genetics , Receptors, Leptin/genetics , Receptors, Pituitary Hormone/genetics , Repressor Proteins/genetics , Retinoid X Receptor alpha/genetics , Sex Factors , Time Factors , Triglycerides/blood , White People/genetics , Young Adult
14.
PLoS One ; 10(10): e0139155, 2015.
Article in English | MEDLINE | ID: mdl-26461262

ABSTRACT

Obesity development during psychotropic treatments represents a major health issue in psychiatry. Melanin-concentrating hormone receptor 2 (MCHR2) is a central receptor involved in energy homeostasis. MCHR2 shares its promoter region with MCHR2-AS1, a long antisense non-coding RNA. The aim of this study was to determine whether tagging single nucleotide polymorphisms (tSNPs) of MCHR2 and MCHR2-AS1 are associated with the body mass index (BMI) in the psychiatric and in the general population. The influence of MCHR2 and MCHR2-AS1 tSNPs on BMI was firstly investigated in a discovery psychiatric sample (n1 = 474). Positive results were tested for replication in two other psychiatric samples (n2 = 164, n3 = 178) and in two population-based samples (CoLaus, n4 = 5409; GIANT, n5 = 113809). In the discovery sample, TT carriers of rs7754794C>T had 1.08 kg/m2 (p = 0.04) lower BMI as compared to C-allele carriers. This observation was replicated in an independent psychiatric sample (-2.18 kg/m2; p = 0.009). The association of rs7754794C>T and BMI seemed stronger in subjects younger than 45 years (median of age). In the population-based sample, a moderate association was observed (-0.17 kg/m2; p = 0.02) among younger individuals (<45y). Interestingly, this association was totally driven by patients meeting lifetime criteria for atypical depression, i.e. major depressive episodes characterized by symptoms such as an increased appetite. Indeed, patients with atypical depression carrying rs7754794-TT had 1.17 kg/m2 (p = 0.04) lower BMI values as compared to C-allele carriers, the effect being stronger in younger individuals (-2.50 kg/m2; p = 0.03; interaction between rs7754794 and age: p-value = 0.08). This study provides new insights on the possible influence of MCHR2 and/or MCHR2-AS1 on obesity in psychiatric patients and on the pathophysiology of atypical depression.


Subject(s)
Body Mass Index , Depression/genetics , Mental Disorders/genetics , Polymorphism, Single Nucleotide/genetics , RNA, Long Noncoding/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Pituitary Hormone/genetics , Adult , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes/genetics , Humans , Middle Aged , Reproducibility of Results , White People/genetics
15.
Gen Comp Endocrinol ; 212: 114-23, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-24662390

ABSTRACT

Melanin-concentrating hormone (MCH) mainly regulates feeding in mammals and pigmentation in teleosts. It acts via two G-protein-coupled receptors, MCH receptor 1 (MCHR1) and MCHR2. Although many studies exploring the MCH system in teleosts and mammals have been carried out, studies on other organisms are limited. In this study, we cloned and characterized four MCHR subtypes from the diploid species Xenopus tropicalis (X-MCHRs; X-MCHR1a, R1b, R2a, and R2b). According to a phylogenetic tree of the X-MCHRs, X-MCHR1a and R2a are close to mammalian MCHRs, while X-MCHR1b and R2b are close to teleostean MCHRs. We previously reported that the G-protein coupling capacity of the MCHR subtypes differed between mammals (R1: Gαi/o and Gαq; R2: Gαq) and teleosts (R1: Gαq; R2: Gαi/o and Gαq) in mammalian cell-based assays. By using Ca(2+) mobilization assays with pertussis toxin in CHO dhfr(-) cells, we found that X-MCHR1a promiscuously coupled to both Gαi/o and Gαq, while X-MCHR1b and R2a exclusively coupled to Gαq. However, no Ca(2+) influx was detected in cells transfected with X-MCHR2b. Reverse transcription-PCR showed that the X-MCHR mRNAs were expressed in various tissues. In particular, both X-MCHR1b and R2b were exclusively found in melanophores of the dorsal skin. In skin pigment migration assays, melanophores were weakly aggregated at low concentrations but dispersed at high concentrations of MCH, suggesting possible interactions between X-MCHR1b and R2b for the regulation of body color. These findings demonstrate that X. tropicalis has four characteristic MCHRs and will be useful for elucidating the nature of MCHR evolution among vertebrates.


Subject(s)
Gene Expression Regulation , Hypothalamic Hormones/genetics , Melanins/genetics , Pituitary Hormones/genetics , Receptors, Pituitary Hormone/genetics , Signal Transduction , Xenopus/genetics , Amino Acid Sequence , Animals , Blotting, Western , CHO Cells , Calcium/metabolism , Cloning, Molecular , Cricetulus , Hypothalamic Hormones/metabolism , Melanins/metabolism , Melanophores/metabolism , Molecular Sequence Data , Phylogeny , Pituitary Hormones/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Pituitary Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Skin/metabolism , Xenopus/growth & development
16.
Behav Brain Res ; 262: 14-20, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24412349

ABSTRACT

Chronic food restriction (FR) and maintenance of low body weight have long been known to increase the rewarding and motor-activating effects of addictive drugs. However, the neurobiological mechanisms through which FR potentiates drug reward remain largely unknown. Melanin-concentrating hormone (MCH) signaling could be one of these mechanisms since this peptide is involved in energy homeostasis and modulates mesolimbic dopaminergic transmission. The purpose of the present study was to test this hypothesis by investigating the impact of FR on amphetamine reward in wild-type (WT) and knockout mice lacking the melanin-concentrating hormone receptor-1 (MCHR1-KO). The rewarding effects of amphetamine (0.75-2.25 mg/kg, i.p.) were measured with the conditioned place preference (CPP) technique. The food of the mice was restricted to maintain their body weight at 80-85% of their free-feeding (FF) weight throughout the entire CPP experiment. Locomotor activity of the animals was recorded during the conditioning sessions. Our results show that locomotion of all the food-restricted mice treated with saline or amphetamine increased over the sessions whatever the genotype. On the place preference test, the amplitude of CPP induced by 0.75 mg/kg amphetamine was higher in food restricted WT mice than in free-fed WT mice and food restricted MCHR1-KO mice. However, FR did not affect amphetamine reward in MCHR1-KO mice. The present results indicate that MCH signaling could be involved in the ability of FR to increase amphetamine-induced CPP.


Subject(s)
Amphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Conditioning, Psychological/drug effects , Food Deprivation , Receptors, Pituitary Hormone/genetics , Reward , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects
17.
Endocrinology ; 155(1): 81-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24169555

ABSTRACT

Melanin-concentrating hormone (MCH) is an orexigenic neuropeptide that is a ligand for two subtypes of MCH receptors, MCHR1 and MCHR2. MCHR1 is universally expressed in mammals ranging from rodents to humans, but the expression of MCHR2 is substantially restricted. In mammals, MCHR2 has been defined in primates as well as other species such as cats and dogs but is not seen in rodents. Although the role of MCHR1 in mediating the actions of MCH on energy balance is clearly defined using mouse models, the role of MCHR2 is harder to characterize because of its limited expression. To determine any potential role of MCHR2 in energy balance, we generated a transgenic MCHR1R2 mouse model, where human MCHR2 is coexpressed in MCHR1-expressing neurons. As shown previously, control wild-type mice expressing only native MCHR1 developed diet-induced obesity when fed a high-fat diet. In contrast, MCHR1R2 mice had lower food intake, leading to their resistance to diet-induced obesity. Furthermore, we showed that MCH action is altered in MCHR1R2 mice. MCH treatment in wild-type mice inhibited the activation of the immediate-early gene c-fos, and coexpression of MCHR2 reduced the inhibitory actions of MCHR1 on this pathway. In conclusion, we developed an experimental animal model that can provide insight into the action of MCHR2 in the central nervous system and suggest that some actions of MCHR2 oppose the endogenous actions of MCHR1.


Subject(s)
Diet , Gene Expression Regulation , Obesity/genetics , Receptors, Pituitary Hormone/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Gene Expression , Glucose/metabolism , Insulin/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Obesity/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Somatostatin/metabolism , Signal Transduction
18.
PLoS One ; 8(12): e83194, 2013.
Article in English | MEDLINE | ID: mdl-24376661

ABSTRACT

BACKGROUND: Melanin-concentrating hormone (MCH), an evolutionarily conserved appetite-regulating neuropeptide, has been recently implicated in the pathogenesis of inflammatory bowel disease (IBD). Expression of MCH is upregulated in inflamed intestinal mucosa in humans with colitis and MCH-deficient mice treated with trinitrobenzene-sulfonic acid (TNBS) develop an attenuated form of colitis compared to wild type animals. Zebrafish have emerged as a new animal model of IBD, although the majority of the reported studies concern zebrafish larvae. Regulation MCH expression in the adult zebrafish intestine remains unknown. METHODS: In the present study we induced enterocolitis in adult zebrafish by intrarectal administration of TNBS. Follow-up included survival analysis, histological assessment of changes in intestinal architecture, and assessment of intestinal infiltration by myeloperoxidase positive cells and cytokine transcript levels. RESULTS: Treatment with TNBS dose-dependently reduced fish survival. This response required the presence of an intact microbiome, since fish pre-treated with vancomycin developed less severe enterocolitis. At 6 hours post-challenge, we detected a significant influx of myeloperoxidase positive cells in the intestine and upregulation of both proinflammatory and anti-inflammatory cytokines. Most importantly, and in analogy to human IBD and TNBS-induced mouse experimental colitis, we found increased intestinal expression of MCH and its receptor in TNBS-treated zebrafish. CONCLUSIONS: Taken together these findings not only establish a model of chemically-induced experimental enterocolitis in adult zebrafish, but point to effects of MCH in intestinal inflammation that are conserved across species.


Subject(s)
Enterocolitis/genetics , Fish Proteins/genetics , Hypothalamic Hormones/genetics , Intestinal Mucosa/metabolism , Melanins/genetics , Pituitary Hormones/genetics , Receptors, Pituitary Hormone/genetics , Administration, Rectal , Animals , Cell Movement , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enterocolitis/chemically induced , Enterocolitis/mortality , Enterocolitis/pathology , Fish Proteins/metabolism , Gene Expression Regulation , Humans , Hypothalamic Hormones/metabolism , Intestines/microbiology , Intestines/pathology , Male , Melanins/metabolism , Microbiota/drug effects , Peroxidase/genetics , Peroxidase/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/metabolism , Survival Analysis , Trinitrobenzenesulfonic Acid , Vancomycin/pharmacology , Zebrafish
19.
J Mol Evol ; 77(5-6): 260-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24193891

ABSTRACT

Mesotocin is a neurohypophyseal hormone found in some non-mammalian vertebrates, including birds, reptiles, and amphibians. In this study, we identified and characterized 18-amino acid duplications in the C-terminal domain of the mesotocin receptor (MTR), specifically found in Turdus thrushes (Aves: Passeriforms: Turdidae). These duplicated elements are located in the distal part of the C-terminal tails of MTR and consist of amino acids that are highly conserved among major vertebrates. Intraspecific polymorphisms in a variable number of tandem duplications are commonly found in East Eurasian Turdus, but not in any other genus of Turdidae. Moreover, the genus Turdus can be further classified into 2 groups according to the presence or absence of a 3-amino acid deletion just adjacent to the putative palmitoylation site in the cytoplasmic C-terminal tail. The phylogeny presented here strongly supports the conspecific group of 4 East Eurasian thrushes (Turdus pallidus, T. chrysolaus, T. obscurus, and T. celaenops). Our findings, therefore, provide a new synapomorphy that can be used for phylogenetic assumptions and shed a light on the history of diversification within Eurasian Turdus clades.


Subject(s)
Passeriformes/genetics , Phylogeny , Polymorphism, Genetic/genetics , Receptors, Pituitary Hormone/genetics , Tandem Repeat Sequences/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , Genotype , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Sex Determination Analysis , Species Specificity
20.
J Clin Oncol ; 31(21): 2654-61, 2013 Jul 20.
Article in English | MEDLINE | ID: mdl-23775959

ABSTRACT

PURPOSE: Previous studies have demonstrated the prognostic importance of the immune microenvironment in follicular lymphoma (FL). To investigate the molecular mechanisms during which tumor-infiltrating T cells (TILs) are altered in the FL microenvironment, we studied highly purified CD4 and CD8 TILs from lymph node biopsies at diagnosis in treatment-naive patients with FL compared with reactive tonsils and the peripheral blood of healthy donors. PATIENTS AND METHODS: Gene expression profiling of highly purified CD4 and CD8 TILs was performed on the Affymetrix platform. Diagnostic tissue microarrays from an independent patient set (n = 172) were used to verify protein expression and analyze any impact of TIL-expressed genes on outcome. Time-lapse imaging was used to assess T-cell motility. RESULTS: The most upregulated genes in both CD4 and CD8 TILs were PMCH, ETV1, and TNFRSF9. PMCH is not expressed in peripheral blood T cells, but expression is highly induced on culture with FL. Both CD4 and CD8 TILs from patients with FL have significantly impaired motility compared with those of healthy TILs from reactive tonsils and this can be induced on healthy T cells by FL cells. During multivariate analysis, a model incorporating the number and location of T cells expressing PMCH, NAMPT, and ETV1 showed prognostic significance for overall survival and for time to transformation. CONCLUSION: We showed altered gene expression in TILs in FL and demonstrated that altering the immune microenvironment in FL affects overall survival and time to transformation in this disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma, Follicular/pathology , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, Pituitary Hormone/biosynthesis , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/immunology , Risk Factors , Survival Analysis , Tissue Array Analysis , Tumor Microenvironment/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...