Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.046
Filter
Add more filters

Publication year range
1.
PLoS Genet ; 18(3): e1010044, 2022 03.
Article in English | MEDLINE | ID: mdl-35271580

ABSTRACT

Congenital idiopathic megaesophagus (CIM) is a gastrointestinal (GI) motility disorder of dogs in which reduced peristaltic activity and dilation of the esophagus prevent the normal transport of food into the stomach. Affected puppies regurgitate meals and water, fail to thrive, and experience complications such as aspiration pneumonia that may necessitate euthanasia. The German shepherd dog (GSD) has the highest disease incidence, indicative of a genetic predisposition. Here, we discover that male GSDs are twice as likely to be affected as females and show that the sex bias is independent of body size. We propose that female endogenous factors (e.g., estrogen) are protective via their role in promoting relaxation of the sphincter between the esophagus and stomach, facilitating food passage. A genome-wide association study for CIM revealed an association on canine chromosome 12 (P-val = 3.12x10-13), with the lead SNPs located upstream or within Melanin-Concentrating Hormone Receptor 2 (MCHR2), a compelling positional candidate gene having a role in appetite, weight, and GI motility. Within the first intron of MCHR2, we identified a 33 bp variable number tandem repeat (VNTR) containing a consensus binding sequence for the T-box family of transcription factors. Across dogs and wolves, the major allele includes two copies of the repeat, whereas the predominant alleles in GSDs have one or three copies. The single-copy allele is strongly associated with CIM (P-val = 1.32x10-17), with homozygosity for this allele posing the most significant risk. Our findings suggest that the number of T-box protein binding motifs may correlate with MCHR2 expression and that an imbalance of melanin-concentrating hormone plays a role in CIM. We describe herein the first genetic factors identified in CIM: sex and a major locus on chromosome 12, which together predict disease state in the GSD with greater than 75% accuracy.


Subject(s)
Esophageal Achalasia , Minisatellite Repeats , Animals , Dogs , Esophageal Achalasia/veterinary , Female , Genome-Wide Association Study , Introns/genetics , Male , Receptors, Pituitary Hormone
2.
Biochem Biophys Res Commun ; 710: 149917, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38604071

ABSTRACT

Melanin-concentrating hormone (MCH) receptor 1 (MCHR1), a G protein-coupled receptor, is poised for interaction with its ligands on the plasma membrane. Analyses of MCHR1 knockout mice suggest that this receptor could be a therapeutic target for the treatment of appetite disorders, glucose metabolism, psychiatric disorders, and inflammation. Binding of MCH to MCHR1 initiates calcium signaling, which is subsequently attenuated through receptor internalization. However, the ultimate destiny of the receptor post-internalization remains unexplored. In this study, we report the extracellular secretion of MCHR1 via exosomes. The recruitment of MCHR1 to exosomes occurs subsequent to its internalization, which is induced by stimulation with the ligand MCH. Although a highly glycosylated form of MCHR1, potentially representing a mature form, is selectively recruited to exosomes, the MCHR1 transferred into other cells does not exhibit functionality. The truncation of MCHR1 at the C-terminus not only impairs its response to MCH but also hinders its recruitment to exosomes. These findings imply that functional MCHR1 could be secreted extracellularly via exosomes, a process that may represent a mechanism for the termination of intracellular MCHR1 signaling.


Subject(s)
Exosomes , Hypothalamic Hormones , Receptors, Pituitary Hormone , Humans , Mice , Animals , Exosomes/metabolism , Receptors, Pituitary Hormone/metabolism , Signal Transduction , Mice, Knockout , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Melanins/metabolism
3.
Bioorg Med Chem Lett ; 84: 129194, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36813053

ABSTRACT

Melanin Concentrating Hormone (MCH) receptor is a G protein-coupled receptor (GPCR) with two subtypes R1 and R2. MCH-R1 is involved in the control of energy homeostasis, feeding behavior and body weight. Many studies have proved that administration of MCH-R1 antagonists significantly reduces food intake and causes weight loss in animal models. Herein, we report the optimization of our previously reported virtual screening hits into novel MCH-R1 ligands with chiral aliphatic nitrogen-containing scaffolds. The activity was improved from the micromolar range of the initial leads to 7 nM. We also disclose the first MCH-R1 ligands based on a diazaspiro[4.5]decane nucleus with sub-micromolar activity. A potent MCH-R1 antagonist with acceptable pharmacokinetic profile could represent a new hope for the management of obesity.


Subject(s)
Receptors, Pituitary Hormone , Animals , Ligands , Receptors, G-Protein-Coupled , Obesity/drug therapy , Melanins
4.
Int J Mol Sci ; 23(7)2022 Mar 30.
Article in English | MEDLINE | ID: mdl-35409167

ABSTRACT

Melanin-concentrating hormone receptor 1 (MCHR1) has been a target for appetite suppressants, which are helpful in treating obesity. However, it is challenging to develop an MCHR1 antagonist because its binding site is similar to that of the human Ether-à-go-go-Related Gene (hERG) channel, whose inhibition may cause cardiotoxicity. Most drugs developed as MCHR1 antagonists have failed in clinical development due to cardiotoxicity caused by hERG inhibition. Machine learning-based prediction models can overcome these difficulties and provide new opportunities for drug discovery. In this study, we identified KRX-104130 with potent MCHR1 antagonistic activity and no cardiotoxicity through virtual screening using two MCHR1 binding affinity prediction models and an hERG-induced cardiotoxicity prediction model. In addition, we explored other possibilities for expanding the new indications for KRX-104130 using a transcriptome-based drug repositioning approach. KRX-104130 increased the expression of low-density lipoprotein receptor (LDLR), which induced cholesterol reduction in the gene expression analysis. This was confirmed by comparison with gene expression in a nonalcoholic steatohepatitis (NASH) patient group. In a NASH mouse model, the administration of KRX-104130 showed a protective effect by reducing hepatic lipid accumulation, liver injury, and histopathological changes, indicating a promising prospect for the therapeutic effect of NASH as a new indication for MCHR1 antagonists.


Subject(s)
Drug Repositioning , Non-alcoholic Fatty Liver Disease , Animals , Cardiotoxicity , Humans , Machine Learning , Mice , Receptors, Pituitary Hormone , Receptors, Somatostatin/metabolism , Transcriptome
5.
Molecules ; 26(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673598

ABSTRACT

Melanin-concentrating hormone (MCH) is a 19 amino acid long peptide found in the brain of animals, including fishes, batrachians, and mammals. MCH is implicated in appetite and/or energy homeostasis. Antagonists at its receptor (MCH-R1) could be major tools (or ultimately drugs) to understand the mechanism of MCH action and to fight the obesity syndrome that is a worldwide societal health problem. Ever since the deorphanisation of the MCH receptor, we cloned, expressed, and characterized the receptor MCH-R1 and started a vast medicinal chemistry program aiming at the discovery of such usable compounds. In the present final work, we describe GPS18169, a pseudopeptide antagonist at the MCH-R1 receptor with an affinity in the nanomolar range and a Ki for its antagonistic effect in the 20 picomolar range. Its metabolic stability is rather ameliorated compared to its initial parent compound, the antagonist S38151. We tested it in an in vivo experiment using high diet mice. GPS18169 was found to be active in limiting the accumulation of adipose tissues and, correlatively, we observed a normalization of the insulin level in the treated animals, while no change in food or water consumption was observed.


Subject(s)
Anti-Obesity Agents/chemistry , Obesity/drug therapy , Receptors, Pituitary Hormone/antagonists & inhibitors , Adipose Tissue/drug effects , Alkynes/chemistry , Aminobutyrates/chemistry , Animals , Anti-Obesity Agents/pharmacology , Appetite/drug effects , Aspartic Acid/chemistry , Disease Models, Animal , Drug Discovery , Glutamic Acid/chemistry , Glycine/analogs & derivatives , Glycine/chemistry , HEK293 Cells , Hepatocytes/drug effects , Homeostasis/drug effects , Humans , Insulin/metabolism , Lactams/chemistry , Male , Mice , Mice, Inbred C57BL , Rats , Structure-Activity Relationship , Tissue Distribution , Triazoles/chemistry
6.
Gen Comp Endocrinol ; 293: 113474, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32240710

ABSTRACT

GPCRs are the largest family of receptors accounting for about 30% of the current drug targets. However, it is difficult to fully elucidate the mechanisms regulating intracellular GPCR signal regulation. It is thus important to consider and investigate GPCRs with respect to endogenous situations. Our group has been investigating GPCRs involved in body color (teleost and amphibian) and eating (vertebrate). Here, I review two independent GPCR systems (heterodimer formation and primary ciliated GPCR) that can be breakthroughs in GPCR research. In teleosts, MCRs form heterodimers, which significantly reduce their affinity for acetylated ligands. In mammals, MCHR1 is localized in the ciliary membrane and shortens the length of the primary cilia through a unique signal from the ciliary membrane. Considering these two new GPCR concepts is expected to advance the overall view of the GPCR system.


Subject(s)
Cilia/metabolism , Protein Multimerization , Receptors, G-Protein-Coupled/metabolism , Receptors, Melanocortin/metabolism , Receptors, Pituitary Hormone/metabolism , Animals , Cell Membrane/metabolism , Humans
7.
Bioorg Med Chem Lett ; 29(24): 126741, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31678007

ABSTRACT

MCH receptor is a G protein-coupled receptor with two subtypes R1 and R2. Many studies have demonstrated the role of MCH-R1 in feeding and energy homeostasis. It has been proven that oral administration of small molecule MCH-R1 antagonists significantly reduces food intake and causes a dose-dependent weight loss. In this study, two ligand-based pharmacophores were developed and validated based on recently published MCH-R1 antagonists with diverse structures. Successful pharmacophores had one hydrogen bond acceptor, one positive ionizable, one ring aromatic and two or three hydrophobic groups. These 3D-QSAR models were used for virtual screening of the ZINC chemical database resulting in the identification of nine compounds with more than 50% displacement of radiolabeled MCH at a 20 µM concentration. Moreover, four of these compounds showed antagonistic activities in Aequorin functional assay, including MH-3 which is the first MCH-R1 antagonist based on a diazaspiro[4.5]decane scaffold. The most active compounds were also docked into our previously published MCH-R1 homology model to gain insights into their binding determinants. These compounds could represent a viable starting scaffold for the design of potent MCH-R1 antagonists with improved pharmacokinetic properties as an effective treatment for obesity.


Subject(s)
Anti-Obesity Agents/therapeutic use , Obesity/drug therapy , Receptors, Pituitary Hormone/antagonists & inhibitors , Anti-Obesity Agents/pharmacology , Humans , Structure-Activity Relationship
8.
Gen Comp Endocrinol ; 281: 91-104, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31121165

ABSTRACT

Two structurally related peptides, arginine vasotocin (AVT) and mesotocin (MT), are reported to regulate many physiological processes, such as anti-diuresis and oviposition in birds, and their actions are likely mediated by four AVT/MT receptors (AVPR1A, AVPR1B, MTR and AVPR2b), which are orthologous/paralogous to human AVPR1A, AVPR1B, OXTR and AVPR2 respectively. However, our knowledge regarding the functions of these avian AVT/MT receptors has been limited. Here, we examined the functionality and expression of these receptors in chickens and investigated the roles of AVT in the anterior pituitary. Our results showed that 1) AVPR1A, AVPR1B and AVPR2b could be preferentially activated by AVT, monitored by cell-based luciferase reporter assays and/or Western blot, indicating that they are AVT-specific receptors (AVPR1A; AVPR1B) or AVT-preferring receptor (AVPR2b) functionally coupled to intracellular calcium, MAPK/ERK and cAMP/PKA signaling pathways. In contrast, MTR could be activated by AVT and MT with similar potencies, indicating that MTR is a receptor common for both peptides; 2) Using qPCR, differential expression of the four receptors was found in chicken tissues including the oviduct and anterior pituitary. In particular, only AVPR1A is abundantly expressed in the uterus, suggesting its involvement in mediating AVT-induced oviposition. 3) In cultured chick pituitary cells, AVT could stimulate ACTH and PRL expression and secretion, an action likely mediated by AVPR1B and/or AVPR1A abundantly expressed in anterior pituitary. Collectively, our data helps to elucidate the roles of AVT/MT in birds, such as the 'oxytocic action' of AVT, which induces uterine muscle contraction during oviposition.


Subject(s)
Oviposition/physiology , Pituitary Gland/metabolism , Prolactin/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Vasopressin/metabolism , Signal Transduction , Vasotocin/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Chickens/metabolism , Ducks , Female , Gene Expression Regulation/drug effects , Models, Biological , Pro-Opiomelanocortin/pharmacology , Prolactin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tissue Distribution , Vasotocin/chemistry
9.
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
10.
Exp Dermatol ; 26(6): 542-543, 2017 06.
Article in English | MEDLINE | ID: mdl-27673728

ABSTRACT

Alopecia areata (AA) is a common hair loss disorder characterized by discrete, well demarcated areas of non-scarring terminal hair alopecia, with the calculated lifetime risk of ~2%. In past decades, linkage and GWA studies have implicated dozens of susceptibility genes/loci that are linked to the development of AA. Fischer et al performed a genome-wide CNV analysis of 585 AA patients and 1,340 controls in a European population. This is the first genome-wide study of CNV to be performed in AA samples, and the association finding in the MCHR2 gene region further underscores the potential role of pigmentation in AA development.


Subject(s)
Alopecia Areata , Receptors, Pituitary Hormone , Cohort Studies , DNA Copy Number Variations , Genome-Wide Association Study , Humans , Pigmentation , Receptors, G-Protein-Coupled
11.
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
12.
Gen Comp Endocrinol ; 253: 44-52, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28842217

ABSTRACT

Primary cilia are specialized microtubule-based organelles. Their importance is highlighted by the gamut of ciliary diseases associated with various syndromes including diabetes and obesity. Primary cilia serve as signaling hubs through selective interactions with ion channels and conventional G-protein-coupled receptors (GPCRs). Melanin-concentrating hormone (MCH) receptor 1 (MCHR1), a key regulator of feeding, is selectively expressed in neuronal primary cilia in distinct regions of the mouse brain. We previously found that MCH acts on ciliary MCHR1 and induces cilia shortening through a Gi/o-dependent Akt pathway with no cell cycle progression. Many factors can participate in cilia length control. However, the mechanisms for how these molecules are relocated and coordinated to activate cilia shortening are poorly understood. In the present study, we investigated the role of cytoskeletal dynamics in regulating MCH-induced cilia shortening using clonal MCHR1-expressing hTERT-RPE1 cells. Pharmacological and biochemical approaches showed that cilia shortening mediated by MCH was associated with increased soluble cytosolic tubulin without changing the total tubulin amount. Enhanced F-actin fiber intensity was also observed in MCH-treated cells. The actions of various pharmacological agents revealed that coordinated actin machinery, especially actin polymerization, was required for MCHR1-mediated cilia shortening. A recent report indicated the existence of actin-regulated machinery for cilia shortening through GPCR agonist-dependent ectosome release. However, our live-cell imaging experiments showed that MCH progressively elicited cilia shortening without exclusion of fluorescence-positive material from the tip. Short cilia phenotypes have been associated with various metabolic disorders. Thus, the present findings may contribute toward better understanding of how the cytoskeleton is involved in the GPCR ligand-triggered cilia shortening with cell mechanical properties that underlies clinical manifestations such as obesity.


Subject(s)
Cilia/metabolism , Cytoskeleton/metabolism , Receptors, Pituitary Hormone/metabolism , Animals , Cell Body/metabolism , Cell Line , Cell-Derived Microparticles/metabolism , Cilia/drug effects , Cytosol/metabolism , Green Fluorescent Proteins/metabolism , Humans , Hypothalamic Hormones/pharmacology , Ligands , Melanins/pharmacology , Mice , Microtubules/metabolism , Models, Biological , Pituitary Hormones/pharmacology , Polymerization , Solubility , Tubulin/metabolism
13.
Dokl Biochem Biophys ; 467(1): 141-4, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27193719

ABSTRACT

We have studied the influence of α-melanocyte-stimulating hormone (α-MSH) on proliferation and early stages of differentiation of human induced pluripotent stem cells (iPSc). We have demonstrated that α-MSH receptor genes are expressed in undifferentiated iPSc. The expression levels of MCR1, MCR2, and MCR3 increased at the embryoid body (EB) formation stage. The formation of neural progenitors was accompanied by elevation of MCR2, MCR3, and MCR4 expression. α-MSH had no effect on EB generation and iPSc proliferation at concentrations ranging from 1 nM to 10 µM. At the same time, α-MSH increased the generation of neural rosettes in human iPSc cultures more than twice.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Induced Pluripotent Stem Cells/physiology , alpha-MSH/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression/physiology , Humans , Neural Stem Cells/physiology , Neurons/physiology , Receptors, Pituitary Hormone/metabolism , alpha-MSH/administration & dosage
14.
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
15.
Neurobiol Dis ; 61: 47-54, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23891728

ABSTRACT

Clinical development of drugs for CNS disorders can be a challenging and risky endeavor. In this article we look at the steps required to move a preclinical candidate compound into clinical development. We use the case study of ALB-127158(a), an MCH1 antagonist for the treatment of obesity via a central mechanism to highlight the steps needed to move into early clinical development. Preclinical studies demonstrated that the compound produced significant weight loss in rodents. Based on the observation that the weight loss was caused by a reduction in food intake it was possible to build measures of ingestive behavior into the early clinical development plan. Single and multiple ascending dose studies were conducted in normal and overweight volunteers. The compound was safe and well tolerated with good PK characteristics. ALB-127158(a) was shown to have some effects on measures of 'hunger' and 'desire to eat', unfortunately these effects only occurred at doses higher than those predicted from the preclinical studies. A subsequent study looking at compound levels in the cerebrospinal fluid (CSF) suggested lower brain exposure than seen in the preclinical models. Based on this data and the limited efficacy observed it was possible to terminate further progression of this compound for obesity before costly long-term weight loss studies were initiated. However, recent reports have demonstrated that MCH acting via MCH1 receptors located on intestinal epithelial cells may be a critical mediator of inflammatory responses within the gastrointestinal (GI) tract. MCH1 receptor antagonists may therefore have a beneficial effect in disorders such as inflammatory bowel disease (IBD). Based on this evidence a peripherally selective MCH1 receptor antagonist such as ALB-127158(a) may be a potential treatment for IBD. This example demonstrates how using data from the preclinical studies is possible to build decision points into an early clinical development plan that will allow early assessment of potential efficacy and allow timely go/no go decisions.


Subject(s)
Anti-Obesity Agents/therapeutic use , Clinical Trials as Topic , Drug Discovery/standards , Drug Evaluation, Preclinical , Indazoles/therapeutic use , Obesity/drug therapy , Pyridones/therapeutic use , Animals , Humans , Male , Rats , Receptors, Pituitary Hormone/antagonists & inhibitors
16.
Gastroenterology ; 144(3): 636-649.e6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23142626

ABSTRACT

BACKGROUND & AIMS: Specific neuronal circuits modulate autonomic outflow to liver and white adipose tissue. Melanin-concentrating hormone (MCH)-deficient mice are hypophagic, lean, and do not develop hepatosteatosis when fed a high-fat diet. Herein, we sought to investigate the role of MCH, an orexigenic neuropeptide specifically expressed in the lateral hypothalamic area, on hepatic and adipocyte metabolism. METHODS: Chronic central administration of MCH and adenoviral vectors increasing MCH signaling were performed in rats and mice. Vagal denervation was performed to assess its effect on liver metabolism. The peripheral effects on lipid metabolism were assessed by real-time polymerase chain reaction and Western blot. RESULTS: We showed that the activation of MCH receptors promotes nonalcoholic fatty liver disease through the parasympathetic nervous system, whereas it increases fat deposition in white adipose tissue via the suppression of sympathetic traffic. These metabolic actions are independent of parallel changes in food intake and energy expenditure. In the liver, MCH triggers lipid accumulation and lipid uptake, with c-Jun N-terminal kinase being an essential player, whereas in adipocytes MCH induces metabolic pathways that promote lipid storage and decreases lipid mobilization. Genetic activation of MCH receptors or infusion of MCH specifically in the lateral hypothalamic area modulated hepatic lipid metabolism, whereas the specific activation of this receptor in the arcuate nucleus affected adipocyte metabolism. CONCLUSIONS: Our findings show that central MCH directly controls hepatic and adipocyte metabolism through different pathways.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Adiposity/physiology , Hypothalamic Area, Lateral/physiology , Hypothalamic Hormones/physiology , Liver/metabolism , Melanins/physiology , Mitogen-Activated Protein Kinase 8/metabolism , Pituitary Hormones/physiology , Adipocytes/drug effects , Adipose Tissue/drug effects , Animals , Eating , Fatty Acids/metabolism , Fatty Liver/metabolism , Fatty Liver/physiopathology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Hormones/administration & dosage , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipogenesis/drug effects , Lipogenesis/physiology , Liver/drug effects , Male , Melanins/administration & dosage , Mice , Non-alcoholic Fatty Liver Disease , Pituitary Hormones/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Hormone/agonists , Receptors, Pituitary Hormone/physiology , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/physiopathology
17.
Brain Inj ; 28(2): 244-51, 2014.
Article in English | MEDLINE | ID: mdl-24295038

ABSTRACT

INTRODUCTION: While it is well established that the brain produces hypothalamic hormones and neuropeptides that influence skeletal metabolism, the impact of traumatic brain injury (TBI) on bone is unknown. Based on the recognition from clinical studies that there is an association between TBI and long-term hypothalamic pituitary dysfunction, it was hypothesized that TBI exerts a negative impact on skeletal growth and maintenance. METHODS: To test the hypothesis, this study employed a repetitive weight drop model for TBI. Four impacts were applied for four consecutive days on 5-week old female C57BL/6 J mice. Bone measurements were taken 2 weeks after the first impact. RESULTS: Bone mineral content (BMC), bone area (B area) and bone mineral density (BMD) in the total body were reduced by 14.5%, 9.8% and 5.2%, respectively, in the impacted vs. control mice. There was a 17.1% reduction in total volumetric BMD (vBMD) and a 4.0% reduction in material vBMD in cortical bone. In trabecular bone, there was a 44.0% reduction in BV/TV. Although there was no change in the cross-sectional bone size, the tibial growth plate and the tibia itself were shortened. CONCLUSION: The repetitive animal TBI model produced an immediate, strong negative impact on bone mass acquisition in young mice.


Subject(s)
Bone and Bones/metabolism , Brain Injuries/metabolism , Osteocalcin/metabolism , Receptors, Pituitary Hormone/metabolism , Animals , Bone Density , Bone Development , Brain Injuries/complications , Brain Injuries/physiopathology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
18.
J Biomol Struct Dyn ; 42(6): 3128-3144, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37216328

ABSTRACT

The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) belongs to the glucagon/secretin family. PACAP interacts with the pituitary adenylate cyclase-activating polypeptide receptor type 1 (PAC1) and vasoactive intestinal peptide receptors 1 and 2 (VPAC1 and VPAC2), exhibiting functions in the immune, endocrine, and nervous systems. This peptide is upregulated in numerous instances of brain injury, acting as a neuroprotective agent. It can also suppress HIV-1 and SARS-CoV-2 viral replication in vitro. This work aimed to identify, in each peptide-receptor system, the most relevant residues for complex stability and interaction energy communication via Molecular Dynamics (MD), Free Energy calculations, and Protein-energy networks, thus revealing in detail the underlying mechanisms of activation of these receptors. Hydrogen bond formation, interaction energies, and computational alanine scanning between PACAP and its receptors showed that His1, Asp3, Arg12, Arg14, and Lys15 are crucial to the peptide's stability. Furthermore, several PACAP interactions with structurally conserved positions deemed necessary in GPCR B1 activation, including Arg2.60, Lys2.67, and Glu7.42, were significant for the peptide's stability within the receptors. According to the protein-energy network, the connection between Asp3 of PACAP and the receptors' conserved Arg2.60 represents a critical energy communication hub in all complexes. Additionally, the ECDs of the receptors were also found to function as energy communication hubs for PACAP. Although the overall binding mode of PACAP in the three receptors was found to be highly conserved, Arg12 and Tyr13 of PACAP were more prominent in complex with PAC1, while Ser2 of PACAP was with VPAC2. The detailed analyses performed in this work pave the way for using PACAP and its receptors as therapeutic targets.Communicated by Ramaswamy H. Sarma.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone , Molecular Dynamics Simulation , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/chemistry , Receptors, Pituitary Hormone/metabolism , Nervous System
19.
Pharmacol Biochem Behav ; 242: 173818, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971471

ABSTRACT

Anxiety disorders are chronic, disabling psychiatric disorders, and there is a growing medical need for the development of novel pharmacotherapeutic agents showing improved efficacy and an improved side effect profile as compared with the currently prescribed anxiolytic drugs. In the course of the search for next-generation anxiolytics, neuropeptide receptors have garnered interest as potential therapeutic targets, underscored by pivotal roles in modulating stress responses and findings from animal studies using pharmacological tools. Among these neuropeptide receptors, the type 1 receptor for melanin-concentrating hormone (MCH1), which has been demonstrated to be involved in an array of physiological processes, including the regulation of stress responses and affective states, has gained attraction as a therapeutic target for drugs used in the treatment of psychiatric disorders, including anxiety disorders. To date, a plethora of MCH1 antagonists have been synthesized, and studies using MCH1 antagonists and genetically manipulated mice lacking MCH1 have revealed that the blockade of MCH1 produces anxiolytic-like effects across diverse rodent paradigms. In addition, MCH1 antagonists have been demonstrated to show a rapid onset of antidepressant-like effects; therefore, they may be effective for conditions commonly encountered in patients with anxiety disorders, which is an advantage for anxiolytic drugs. Notably, MCH1 antagonists have not manifested the undesirable side effects observed with the currently prescribed anxiolytics. All these preclinical findings testify to the potential of MCH1 antagonists as novel anxiolytics. Although there are still issues that need to be resolved prior to the initiation of clinical trials, such as elucidating the precise neuronal mechanisms underlying their anxiolytic effects and exploring pertinent biomarkers that can be used in clinical trials, MCH1 blockade appears to be an attractive way to tackle anxiety disorders.


Subject(s)
Anti-Anxiety Agents , Anxiety Disorders , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Humans , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Receptors, Pituitary Hormone/antagonists & inhibitors , Receptors, Pituitary Hormone/metabolism , Mice
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL