1
|
Brar HK, Chen E, Chang F, Lu SA, Longowal DK, Moon KM, Foster LJ, Reiner N, Nandan D. Leishmania regulates host YY1: Comparative proteomic analysis identifies infection modulated YY1 dependent proteins. PLoS One 2025; 20:e0323227. [PMID: 40373059 DOI: 10.1371/journal.pone.0323227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/04/2025] [Indexed: 05/17/2025] Open
Abstract
The protein Yin-Yang 1 (YY1) is a ubiquitous multifunctional transcription factor. Interestingly, there are several cellular functions controlled by YY1 that could play a role in Leishmania pathogenesis. Leishmaniasis is a human disease caused by protozoan parasites of the genus Leishmania. This study examined the potential role of macrophage YY1 in promoting Leishmania intracellular survival. Deliberate knockdown of YY1 resulted in attenuated survival of Leishmania in infected macrophages, suggesting a role of YY1 in Leishmania persistence. Biochemical fractionation studies revealed Leishmania infection caused redistribution of YY1 to the cytoplasm from the nucleus where it is primarily located. Inhibition of nuclear transport by leptomycin B attenuates infection-mediated YY1 redistribution and reduces Leishmania survival. This suggests that Leishmania induces the translocation of YY1 from the nucleus to the cytoplasm of infected cells, where it may regulate host molecules to favour parasite survival. A label-free quantitative whole proteome approach showed that the expression of a large number of macrophage proteins was dependent on the YY1 level. Interestingly, several of these proteins were modulated in Leishmania-infected cells, revealing YY1-dependent host response and suggesting their potential role in Leishmania pathogenesis. Together, this study identifies YY1 as a novel virulence factor that promotes Leishmania survival inside host macrophages.
Collapse
Affiliation(s)
- Harsimran Kaur Brar
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eleanor Chen
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fabian Chang
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shawna Angel Lu
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dilraj Kaur Longowal
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neil Reiner
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Devki Nandan
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Park JY, Wu C, Basu S, McGue M, Pan W. Adaptive SNP-Set Association Testing in Generalized Linear Mixed Models with Application to Family Studies. Behav Genet 2018; 48:55-66. [PMID: 29150721 PMCID: PMC5754233 DOI: 10.1007/s10519-017-9883-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
In genome-wide association studies (GWAS), it has been increasingly recognized that, as a complementary approach to standard single SNP analyses, it may be beneficial to analyze a group of functionally related SNPs together. Among the existent population-based SNP-set association tests, two adaptive tests, the aSPU test and the aSPUpath test, offer a powerful and general approach at the gene- and pathway-levels by data-adaptively combining the results across multiple SNPs (and genes) such that high statistical power can be maintained across a wide range of scenarios. We extend the aSPU and the aSPUpath test to familial data under the framework of the generalized linear mixed models (GLMMs), which can take account of both subject relatedness and possible population structure. As in population-based GWAS, the proposed aSPU and aSPUpath tests require only fitting a single and common GLMM (under the null hypothesis) for all the SNPs, thus are computationally efficient and feasible for large GWAS data. We illustrate our approaches in identifying genes and pathways associated with alcohol dependence in the Minnesota Twin Family Study. The aSPU test detected a gene associated with the trait, in contrast to none by the standard single SNP analysis. Our aSPU test also controlled Type I errors satisfactorily in a small simulation study. We provide R code to conduct the aSPU and aSPUpath tests for familial and other correlated data.
Collapse
Affiliation(s)
- Jun Young Park
- Division of Biostatistics, University of Minnesota, A460 Mayo Building, MMC 303, 420 Delaware St. SE, Minneapolis, MN, 55455, USA
| | - Chong Wu
- Division of Biostatistics, University of Minnesota, A460 Mayo Building, MMC 303, 420 Delaware St. SE, Minneapolis, MN, 55455, USA
| | - Saonli Basu
- Division of Biostatistics, University of Minnesota, A460 Mayo Building, MMC 303, 420 Delaware St. SE, Minneapolis, MN, 55455, USA
| | - Matt McGue
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, A460 Mayo Building, MMC 303, 420 Delaware St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
3
|
Hagar JM, Macht VA, Wilson SP, Fadel JR. Upregulation of orexin/hypocretin expression in aged rats: Effects on feeding latency and neurotransmission in the insular cortex. Neuroscience 2017; 350:124-132. [PMID: 28344067 DOI: 10.1016/j.neuroscience.2017.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
Abstract
Aging is associated with changes in numerous homeostatic functions, such as food intake, that are thought to be mediated by the hypothalamus. Orexin/hypocretin neurons of the hypothalamus regulate several physiological functions, including feeding, sleep and wakefulness. Evidence from both clinical and animal studies supports the notion that aging is associated with loss or dysregulation of the orexin system. Here, we used virus-mediated gene transfer to manipulate expression of orexin peptides in young and aged rats and examined behavioral and neurochemical correlates of food intake in these animals. Aged rats showed slower feeding latencies when presented with palatable food compared to young control rats, and these deficits were ameliorated by upregulation of orexin expression. Similarly, young animals treated with a virus designed to decrease preproorexin expression showed longer feeding latencies reminiscent of aged control rats. Feeding was also associated with increased acetylcholine, glutamate and GABA efflux in insular cortex of young control animals. Orexin upregulation did not restore deficits in feeding-elicited release of these neurotransmitters in aged rats, but did enhance basal neurotransmitter levels which may have contributed to the behavioral correlates of these genetic manipulations. These studies demonstrate that age-related deficits in behavioral and neurochemical measures of feeding are likely to be mediated, in part, by the orexin system. Because these same neurotransmitter systems have been shown to underlie orexin effects on cognition, treatments which increase orexin function may have potential for improving both physiological and cognitive manifestations of certain age-related disorders.
Collapse
Affiliation(s)
- Janel M Hagar
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Victoria A Macht
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA; Department of Psychology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Steven P Wilson
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - James R Fadel
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| |
Collapse
|
4
|
Understanding the comparative molecular field analysis (CoMFA) in terms of molecular quantum similarity and DFT-based reactivity descriptors. J Mol Model 2015; 21:156. [DOI: 10.1007/s00894-015-2690-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
|
5
|
Faster, better, stronger: towards new antidepressant therapeutic strategies. Eur J Pharmacol 2014; 753:32-50. [PMID: 25092200 DOI: 10.1016/j.ejphar.2014.07.046] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 05/28/2014] [Accepted: 07/24/2014] [Indexed: 12/26/2022]
Abstract
Major depression is a highly prevalent disorder and is predicted to be the second leading cause of disease burden by 2020. Although many antidepressant drugs are currently available, they are far from optimal. Approximately 50% of patients do not respond to initial first line antidepressant treatment, while approximately one third fail to achieve remission following several pharmacological interventions. Furthermore, several weeks or months of treatment are often required before clinical improvement, if any, is reported. Moreover, most of the commonly used antidepressants have been primarily designed to increase synaptic availability of serotonin and/or noradrenaline and although they are of therapeutic benefit to many patients, it is clear that other therapeutic targets are required if we are going to improve the response and remission rates. It is clear that more effective, rapid-acting antidepressants with novel mechanisms of action are required. The purpose of this review is to outline the current strategies that are being taken in both preclinical and clinical settings for identifying superior antidepressant drugs. The realisation that ketamine has rapid antidepressant-like effects in treatment resistant patients has reenergised the field. Further, developing an understanding of the mechanisms underlying the rapid antidepressant effects in treatment-resistant patients by drugs such as ketamine may uncover novel therapeutic targets that can be exploited to meet the Olympian challenge of developing faster, better and stronger antidepressant drugs.
Collapse
|
6
|
The melanin-concentrating hormone receptors: neuronal and non-neuronal functions. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2014; 4:S31-6. [PMID: 27152164 DOI: 10.1038/ijosup.2014.9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Melanin-concentrating hormone (MCH) is a cyclic peptide highly conserved in vertebrates and was originally identified as a skin-paling factor in Teleosts. In fishes, MCH also participates in the regulation of the stress-response and feeding behaviour. Mammalian MCH is a hypothalamic neuropeptide that displays multiple functions, mostly controlling feeding behaviour and energy homeostasis. Transgenic mouse models and pharmacological studies have shown the importance of the MCH system as a potential target in the treatment of appetite disorders and obesity as well as anxiety and psychiatric diseases. Two G-protein-coupled receptors (GPCRs) binding MCH have been characterized so far. The first, named MCH-R1 and also called SLC1, was identified through reverse pharmacology strategies by several groups as a cognate receptor of MCH. This receptor is expressed at high levels in many brain areas of rodents and primates and is also expressed in peripheral organs, albeit at a lower rate. A second receptor, designated MCH-R2, exhibited 38% identity to MCH-R1 and was identified by sequence analysis of the human genome. Interestingly, although MCH-R2 orthologues were also found in fishes, dogs, ferrets and non-human primates, this MCH receptor gene appeared either lacking or non-functional in rodents and lagomorphs. Both receptors are class I GPCRs, whose main roles are to mediate the actions of peptides and neurotransmitters in the central nervous system. However, examples of action of MCH on neuronal and non-neuronal cells are emerging that illustrate novel MCH functions. In particular, the functionality of endogenously expressed MCH-R1 has been explored in human neuroblastoma cells, SK-N-SH and SH-SY5Y cells, and in non-neuronal cell types such as the ependymocytes. Indeed, we have identified mitogen-activated protein kinase (MAPK)-dependent or calcium-dependent signalling cascades that ultimately contributed to neurite outgrowth in neuroblastoma cells or to modulation of ciliary beating in ependymal cells. The putative role of MCH on cellular shaping and plasticity on one side and volume transmission on the other must be now considered.
Collapse
|
7
|
MacNeil DJ. The role of melanin-concentrating hormone and its receptors in energy homeostasis. Front Endocrinol (Lausanne) 2013; 4:49. [PMID: 23626585 PMCID: PMC3631741 DOI: 10.3389/fendo.2013.00049] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 04/09/2013] [Indexed: 01/25/2023] Open
Abstract
Extensive studies in rodents with melanin-concentrating hormone (MCH) have demonstrated that the neuropeptide hormone is a potent orexigen. Acutely, MCH causes an increase in food intake, while chronically it leads to increased weight gain, primarily as an increase in fat mass. Multiple knockout mice models have confirmed the importance of MCH in modulating energy homeostasis. Animals lacking MCH, MCH-containing neurons, or the MCH receptor all are resistant to diet-induced obesity. These genetic and pharmacologic studies have prompted a large effort to identify potent and selective MCH receptor antagonists, initially as tool compounds to probe pharmacology in models of obesity, with an ultimate goal to identify novel anti-obesity drugs. In animal models, MCH antagonists have consistently shown efficacy in reducing food intake acutely and inhibiting body-weight gain when given chronically. Five compounds have proceeded into clinical testing. Although they were reported as well-tolerated, none has advanced to long-term efficacy and safety studies.
Collapse
Affiliation(s)
- Douglas J. MacNeil
- Department of In Vitro Pharmacology, Merck Research LaboratoriesKenilworth, NJ, USA
- *Correspondence: Douglas J. MacNeil, Department of In Vitro Pharmacology, Merck Research Laboratories, K15-3-309D, 2015 Galloping Hill Road, Kenilworth, NJ 07033, USA. e-mail:
| |
Collapse
|
8
|
Högberg T, Frimurer TM, Sasmal PK. Melanin concentrating hormone receptor 1 (MCHR1) antagonists—Still a viable approach for obesity treatment? Bioorg Med Chem Lett 2012; 22:6039-47. [DOI: 10.1016/j.bmcl.2012.08.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 07/31/2012] [Accepted: 08/02/2012] [Indexed: 12/12/2022]
|
9
|
Sasmal S, Balaji G, Kanna Reddy HR, Balasubrahmanyam D, Srinivas G, Kyasa SK, Sasmal PK, Khanna I, Talwar R, Suresh J, Jadhav VP, Muzeeb S, Shashikumar D, Harinder Reddy K, Sebastian VJ, Frimurer TM, Rist Ø, Elster L, Högberg T. Design and optimization of quinazoline derivatives as melanin concentrating hormone receptor 1 (MCHR1) antagonists. Bioorg Med Chem Lett 2012; 22:3157-62. [PMID: 22487182 DOI: 10.1016/j.bmcl.2012.03.050] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/11/2012] [Accepted: 03/13/2012] [Indexed: 11/24/2022]
Abstract
Melanin concentrating hormone (MCH) is an important mediator of energy homeostasis and plays a role in metabolic and CNS disorders. The modeling-supported design, synthesis and multi-parameter optimization (biological activity, solubility, metabolic stability, hERG) of novel quinazoline derivatives as MCHR1 antagonists are described. The in vivo proof of principle for weight loss with a lead compound from this series is exemplified. Clusters of refined hMCHR1 homology models derived from the X-ray structure of the β2-adrenergic receptor, including extracellular loops, were developed and used to guide the design.
Collapse
Affiliation(s)
- Sanjita Sasmal
- Discovery Research, Dr. Reddy's Laboratories Ltd, Bollaram Road, Miyapur, Hyderabad 500049, India.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Sasmal S, Balasubrahmanyam D, Kanna Reddy HR, Balaji G, Srinivas G, Cheera S, Abbineni C, Sasmal PK, Khanna I, Sebastian V, Jadhav VP, Singh MP, Talwar R, Suresh J, Shashikumar D, Harinder Reddy K, Sihorkar V, Frimurer TM, Rist Ø, Elster L, Högberg T. Design and optimization of quinazoline derivatives as melanin concentrating hormone receptor 1 (MCHR1) antagonists: Part 2. Bioorg Med Chem Lett 2012; 22:3163-7. [DOI: 10.1016/j.bmcl.2012.03.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 03/11/2012] [Accepted: 03/13/2012] [Indexed: 10/28/2022]
|
11
|
Kasai S, Kamata M, Masada S, Kunitomo J, Kamaura M, Okawa T, Takami K, Ogino H, Nakano Y, Ashina S, Watanabe K, Kaisho T, Imai YN, Ryu S, Nakayama M, Nagisa Y, Takekawa S, Kato K, Murata T, Suzuki N, Ishihara Y. Synthesis, structure-activity relationship, and pharmacological studies of novel melanin-concentrating hormone receptor 1 antagonists 3-aminomethylquinolines: reducing human ether-a-go-go-related gene (hERG) associated liabilities. J Med Chem 2012; 55:4336-51. [PMID: 22490048 DOI: 10.1021/jm300167z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recently, we discovered 3-aminomethylquinoline derivative 1, a selective, highly potent, centrally acting, and orally bioavailable human MCH receptor 1 (hMCHR1) antagonist, that inhibited food intake in F344 rats with diet-induced obesity (DIO). Subsequent investigation of 1 was discontinued because 1 showed potent hERG K(+) channel inhibition in a patch-clamp study. To decrease hERG K(+) channel inhibition, experiments with ligand-based drug designs based on 1 and a docking study were conducted. Replacement of the terminal p-fluorophenyl group with a cyclopropylmethoxy group, methyl group introduction on the benzylic carbon at the 3-position of the quinoline core, and employment of a [2-(acetylamino)ethyl]amino group as the amine portion eliminated hERG K(+) channel inhibitory activity in a patch-clamp study, leading to the discovery of N-{3-[(1R)-1-{[2-(acetylamino)ethyl]amino}ethyl]-8-methylquinolin-7-yl}-4-(cyclopropylmethoxy)benzamide (R)-10h. The compound (R)-10h showed potent inhibitory activity against hMCHR1 and dose-dependently suppressed food intake in a 2-day study on DIO-F344 rats. Furthermore, practical chiral synthesis of (R)-10h was performed to determine the molecule's absolute configuration.
Collapse
Affiliation(s)
- Shizuo Kasai
- Pharmaceutical Research Division, Takeda Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Della-Zuana O, Audinot V, Levenez V, Ktorza A, Presse F, Nahon JL, Boutin JA. Peripheral injections of melanin-concentrating hormone receptor 1 antagonist S38151 decrease food intake and body weight in rodent obesity models. Front Endocrinol (Lausanne) 2012; 3:160. [PMID: 23267345 PMCID: PMC3527734 DOI: 10.3389/fendo.2012.00160] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 11/26/2012] [Indexed: 12/02/2022] Open
Abstract
The compound S38151 is a nanomolar antagonist that acts at the melanin-concentrating hormone receptor 1 (MCH(1)). S38151 is more stable than its purely peptide counterpart, essentially because of the blockade of its N-terminus. Therefore, its action on various models of obesity was studied. Acute intra-cerebroventricular (i.c.v.) administration of S38151 in wild-type rats counteracted the effect of the stable precursor of melanin-concentrating hormone (MCH), NEI-MCH, in a dose-dependent manner (from 0.5 to 50 nmol/kg). In genetically obese Zucker fa/fa rats, daily i.c.v. administration of S38151 induced dose-dependent (5, 10, and 20 nmol/kg) inhibition of food intake, water intake, and body weight gain, as well as increased motility (maximal effect observed at 20 nmol/kg). In Zucker fa/fa rats, intraperitoneal injection of S38151 (30 mg/kg) induced complete inhibition of food consumption within 1 h. Daily intraperitoneal injection of S38151 (10 and 30 mg/kg) into genetically obese ob/ob mice or diet-induced obese mice is able to limit body weight gain. Furthermore, S38151 administration (10 and 30 mg/kg) does not affect food intake, water intake, or body weight gain in MCHR1-deleted mice, demonstrating that its effects are linked to its interaction with MCH(1). These results validate MCH(1) as a target of interest in obesity. S38151 cannot progress to the clinical phase because it is still too poorly stable in vivo.
Collapse
Affiliation(s)
- Odile Della-Zuana
- Maladies Métaboliques, Institut de Recherches SERVIERSuresnes, France
| | - Valérie Audinot
- Biotechnologie, Pharmacologie Moléculaire et Cellulaire, Institut de Recherches SERVIERCroissy-sur-Seine, France
| | - Viviane Levenez
- Maladies Métaboliques, Institut de Recherches SERVIERSuresnes, France
| | - Alain Ktorza
- Maladies Métaboliques, Institut de Recherches SERVIERSuresnes, France
| | - Françoise Presse
- Genomics and Evolution in Neuroendocrinology, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Centre National de la Recherche ScientifiqueValbonne, France
- Genomics and Evolution in Neuroendocrinology, Université de Nice Sophia AntipolisNice, France
| | - Jean-Louis Nahon
- Genomics and Evolution in Neuroendocrinology, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Centre National de la Recherche ScientifiqueValbonne, France
- Genomics and Evolution in Neuroendocrinology, Université de Nice Sophia AntipolisNice, France
| | - Jean A. Boutin
- Biotechnologie, Pharmacologie Moléculaire et Cellulaire, Institut de Recherches SERVIERCroissy-sur-Seine, France
- *Correspondence: Jean A. Boutin, Biotechnologie, Pharmacologie Moléculaire et Cellulaire, Institut de Recherches SERVIER, 125 chemin de Ronde, 78290 Croissy-sur-Seine, France. e-mail:
| |
Collapse
|
13
|
Lim CJ, Kim SH, Lee BH, Oh KS, Yi KY. 4-Arylphthalazin-1(2H)-one derivatives as potent antagonists of the melanin concentrating hormone receptor 1 (MCH-R1). Bioorg Med Chem Lett 2012; 22:427-30. [DOI: 10.1016/j.bmcl.2011.10.111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 10/28/2011] [Accepted: 10/31/2011] [Indexed: 11/29/2022]
|
14
|
Sasmal PK, Sasmal S, Abbineni C, Venkatesham B, Rao PT, Roshaiah M, Khanna I, Sebastian VJ, Suresh J, Singh MP, Talwar R, Shashikumar D, Reddy KH, Frimurer TM, Rist Ø, Elster L, Högberg T. Synthesis and SAR studies of benzimidazole derivatives as melanin concentrating hormone receptor 1 (MCHR1) antagonists: Focus to detune hERG inhibition. MEDCHEMCOMM 2011. [DOI: 10.1039/c1md00015b] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
15
|
Discovery of novel, orally available benzimidazoles as melanin concentrating hormone receptor 1 (MCHR1) antagonists. Bioorg Med Chem Lett 2010; 20:5443-8. [DOI: 10.1016/j.bmcl.2010.07.086] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 07/20/2010] [Accepted: 07/22/2010] [Indexed: 11/19/2022]
|
16
|
Griffond B, Risold PY. MCH and feeding behavior-interaction with peptidic network. Peptides 2009; 30:2045-51. [PMID: 19619600 DOI: 10.1016/j.peptides.2009.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/17/2009] [Accepted: 07/09/2009] [Indexed: 12/20/2022]
Abstract
Numerous works associate the MCH peptide, and the hypothalamic neurons that produce it, to the feeding behavior and energy homeostasis. It is commonly admitted that MCH is an orexigenic peptide, and MCH neurons could be under the control of arcuate NPY and POMC neurons. However, the literature data is not always concordant. In particular questions about the intrahypothalamic circuit involving other neuropeptides and about the mechanisms through which MCH could act are not yet clearly answered. For example, which receptors mediate a MCH response to NPY or alpha-MSH, does MCH act alone, is there any local anatomical organization within the tuberal LHA? A review of the current literature is then needed to help focus attention on these unresolved and often neglected issues.
Collapse
Affiliation(s)
- B Griffond
- Université de Franche-Comté, Besançon, France
| | | |
Collapse
|
17
|
Cotta-Grand N, Rovère C, Guyon A, Cervantes A, Brau F, Nahon JL. Melanin-concentrating hormone induces neurite outgrowth in human neuroblastoma SH-SY5Y cells through p53 and MAPKinase signaling pathways. Peptides 2009; 30:2014-24. [PMID: 19540893 DOI: 10.1016/j.peptides.2009.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 06/05/2009] [Accepted: 06/11/2009] [Indexed: 01/08/2023]
Abstract
Melanin-concentrating hormone (MCH) peptide plays a major role in energy homeostasis regulation. Little is known about cellular functions engaged by endogenous MCH receptor (MCH-R1). Here, MCH-R1 mRNA and cognate protein were found expressed in human neuroblastoma SH-SY5Y cells. Electrophysiological experiments demonstrated that MCH modulated K(+) currents, an effect depending upon the time of cellular growth. MCH treatments induced a transient phosphorylation of MAPKinases, abolished by PD98059, and partially blocked by PTX, suggesting a Galphai/Galphao protein contribution. MCH stimulated expression and likely nuclear localization of phosphorylated p53 proteins, an effect fully dependent upon MAPKinase activities. MCH treatment also increased phosphorylation of Elk-1 and up-regulated Egr-1, two transcriptional factors targeted by the MAPKinase pathway. Finally, MCH provoked neurite outgrowth after 24h-treatment of neuroblastoma cells. This effect and transcriptional factors activation were partly prevented by PD98059. Collectively, our results provide the first evidence for a role of MCH in neuronal differentiation of endogenously MCH-R1-expressing cells via non-exclusive MAPKinase and p53 signaling pathways.
Collapse
Affiliation(s)
- Natacha Cotta-Grand
- The Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique and Université Nice-Sophia Antipolis, Valbonne, France
| | | | | | | | | | | |
Collapse
|
18
|
Vaudry H, Nahon JL. Melanin-concentrating hormone. Editorial. Peptides 2009; 30:1965-6. [PMID: 19765628 DOI: 10.1016/j.peptides.2009.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 09/08/2009] [Indexed: 11/19/2022]
|
19
|
Ito M, Ishihara A, Gomori A, Egashira S, Matsushita H, Mashiko S, Ito J, Ito M, Nakase K, Haga Y, Iwaasa H, Suzuki T, Ohtake N, Moriya M, Sato N, MacNeil DJ, Takenaga N, Tokita S, Kanatani A. Melanin-concentrating hormone 1-receptor antagonist suppresses body weight gain correlated with high receptor occupancy levels in diet-induced obesity mice. Eur J Pharmacol 2009; 624:77-83. [PMID: 19836369 DOI: 10.1016/j.ejphar.2009.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 07/18/2009] [Accepted: 10/06/2009] [Indexed: 11/25/2022]
Abstract
Melanin-concentrating hormone (MCH), which is a neuropeptide expressed in the hypothalamus of the brain, is involved in regulating feeding behavior and energy homeostasis via the MCH(1) receptor in rodents. It is widely considered that MCH(1) receptor antagonists are worthy of development for medical treatment of obesity. Here we report on the development of an ex vivo receptor occupancy assay using a new radiolabeled MCH(1) receptor antagonist, [(35)S]-compound D. An MCH(1) receptor antagonist inhibited the binding of [(35)S]-compound D to brain slices in a dose-dependent manner. The result showed a good correlation between the receptor occupancy levels and plasma or brain levels of the MCH(1) receptor antagonist, suggesting that the ex vivo receptor binding assay using this radioligand is practical. Quantitative analysis in diet-induced obese mice showed that the efficacy of body weight reduction correlated with the receptor occupancy levels at 24h. Furthermore, more than 90% occupancy levels of MCH(1) receptor antagonists during 24h post-dosing are required for potent efficacy on body weight reduction. The present occupancy assay could be a useful pharmacodynamic marker to quantitatively estimate anti-obese efficacy, and would accelerate the development of MCH(1) receptor antagonists for treatment of obesity.
Collapse
Affiliation(s)
- Masahiko Ito
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hegde LG, Ping XL, Jochnowitz N, Craig DA. The role of melanin-concentrating hormone-1 receptors in the voiding reflex in rats. J Pharmacol Exp Ther 2009; 328:165-73. [PMID: 18849359 DOI: 10.1124/jpet.108.143495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
We have used the selective melanin-concentrating hormone-1 (MCH(1)) receptor antagonist SNAP 7941 [((+)-methyl (4S)-3-{[(3-{4-[3-(acetylamino)phenyl]-1-piperidinyl}propyl) amino]carbonyl}-4-(3,4-difluorophenyl)-6-(methoxymethyl)-2-oxo-1,2,3,4-tetrahydro-5-pyrimidinecarboxylate hydrochloride)] to investigate the role of the hypothalamic neuropeptide MCH in the control of voiding in rats. Intravenous administration of SNAP 7941 (3 and 10 mg/kg i.v.) produced dose-related inhibition of rhythmic, distension-induced voiding contractions in anesthetized rats. In conscious rats in which repeated voiding cycles were evoked by continuous slow transvesicular infusion of saline, intragastric SNAP 7941 [0.03-1 mg/kg intragastrically (i.g.)] produced sustained increases in infusion capacity (maximum = 220% basal), comparable with the effects of the 5-hydroxytryptamine(1A) antagonist WAY 100635 (N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinyl-cyclohexanecarboxamide maleate salt), and the muscarinic antagonist, oxybutynin (4-diethylaminobut-2-ynyl 2-cyclohexyl-2-hydroxy-2-phenylacetate hydrochloride). SNAP 7941 produced similar results when administered at a low dose (0.01 nmol) into the lateral ventricle (intracerebroventricular). The opposite effect was produced when MCH (20 nmol) was delivered intracerebroventricularly, resulting in a 34% decrease in apparent bladder capacity with increased urinary frequency. The effect of MCH was blocked by the prior intragastric administration of SNAP 7941 (0.1 mg/kg), but oxybutynin (1 mg/kg) was ineffective. Finally, in conscious spontaneously hypertensive rats, SNAP 7941 (0.1 mg/kg i.g.) produced a 31% reduction in micturition frequency, accompanied by a 36% increase in bladder capacity, with no effect on total volume voided over 6 h. The data indicate that MCH acts via MCH(1) receptors within the CNS to modulate the voiding reflex in rats. The striking effects of the MCH(1) antagonist SNAP 7941 to increase bladder capacity and reduce voiding frequency indicate that MCH(1) antagonists may offer a potential novel approach for treating overactive bladder syndrome.
Collapse
Affiliation(s)
- Laxminarayan G Hegde
- Department of Target Discovery and Assessment, Lundbeck Research USA, Inc., 215 College Road, Paramus, NJ 07652-1431, USA
| | | | | | | |
Collapse
|
21
|
The melanin-concentrating hormone1 receptor antagonists, SNAP-7941 and GW3430, enhance social recognition and dialysate levels of acetylcholine in the frontal cortex of rats. Int J Neuropsychopharmacol 2008; 11:1105-22. [PMID: 18466669 DOI: 10.1017/s1461145708008894] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Melanin-concentrating hormone (MCH)1 receptors are widely expressed in limbic structures and cortex. Their inactivation is associated with anxiolytic and antidepressive properties but little information is available concerning cognition. This issue was addressed using the selective antagonists, SNAP-7941 and GW3430, in a social recognition paradigm in rats. The muscarinic blocker, scopolamine (1.25 mg/kg s.c.), reduced social recognition, an action dose-dependently blocked by SNAP-7941 and GW3430 (0.63-10.0 and 20.0-80.0 mg/kg i.p., respectively) which did not themselves display amnesic properties. Further, in a protocol where a spontaneous deficit was induced by a prolonged inter-session delay, SNAP-7941 and GW3430 dose-dependently enhanced social recognition. In dialysis studies, SNAP-7941 (0.63-40.0 mg/kg i.p.) and GW3430 (10.0-40.0 mg/kg i.p.) elevated extracellular levels of acetylcholine (ACh) in the frontal cortex (FCX) of freely moving rats. The SNAP-7941 effect was specific, as it did not increase levels of ACh in ventral and dorsal hippocampus: moreover, it did not modify levels of noradrenaline, dopamine, serotonin and glutamate in FCX. Active doses of SNAP-7941 and GW3430 corresponded to doses (2.5-40.0 and 10.0-80.0 mg/kg i.p., respectively) exerting anxiolytic properties in Vogel conflict and ultrasonic vocalization tests, and antidepressant actions in forced swim, isolation-induced aggression and marble-burying procedures. In contrast to SNAP-7941 and GW3430, the benzodiazepine, diazepam, decreased social recognition and dialysate levels of ACh, while the tricyclic, imipramine, reduced social recognition and failed to enhance cholinergic transmission. In conclusion, at anxiolytic and antidepressant doses, SNAP-7941 and GW3430 improve social recognition and elevate extracellular ACh levels in FCX. This profile differentiates MCH1 receptor antagonists from conventional anxiolytic and antidepressant agents.
Collapse
|
22
|
Kerman IA, Bernard R, Rosenthal D, Beals J, Akil H, Watson SJ. Distinct populations of presympathetic-premotor neurons express orexin or melanin-concentrating hormone in the rat lateral hypothalamus. J Comp Neurol 2008; 505:586-601. [PMID: 17924541 DOI: 10.1002/cne.21511] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Orexin and melanin-concentrating hormone (MCH) have been implicated in mediating a variety of different behaviors. These include sleep and wakefulness, locomotion, ingestive behaviors, and fight-or-flight response, as well as anxiety- and panic-like behaviors in rodents. Despite such diversity, all these processes require coordinated recruitment of the autonomic and somatomotor efferents. We have previously mapped the locations of presympathetic-premotor neurons (PSPMNs) in the rat brain. These putative dual-function neurons send trans-synaptic projections to somatomotor and sympathetic targets and likely participate in somatomotor-sympathetic integration. A significant portion of these neurons is found within the dorsomedial (DMH) and lateral hypothalamus (LH), areas of the brain that contain MCH- and orexin- synthesizing neurons in the central nervous system. Thus, we hypothesized that hypothalamic PSPMNs utilize MCH or orexin as their neurotransmitter. To test this hypothesis, we identified PSPMNs by using recombinant strains of the pseudorabies virus (PRV) for trans-synaptic tract tracing. PRV-152, a strain that expresses enhanced green fluorescent protein, was injected into sympathectomized gastrocnemius muscle, whereas PRV-BaBlu, which expresses beta-galactosidase, was injected into the adrenal gland in the same animals. By using immunofluorescent methods, we determined whether co-infected neurons express MCH or orexin. Our findings demonstrate that PSPMNs synthesizing either MCH or orexin are present within LH, where they form two separate populations. PSPMNs located around the fornix express orexin, whereas those located around the cerebral peduncle are more likely to express MCH. These two clusters of PSPMNs within LH likely play distinct functional roles in autonomic homeostasis and stress coping mechanisms.
Collapse
Affiliation(s)
- Ilan A Kerman
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Design and synthesis of novel hydantoin-containing melanin-concentrating hormone receptor antagonists. Bioorg Med Chem Lett 2007; 17:3754-9. [PMID: 17448659 DOI: 10.1016/j.bmcl.2007.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2007] [Revised: 03/31/2007] [Accepted: 04/05/2007] [Indexed: 11/16/2022]
Abstract
We report here new chemical series acting as antagonists of melanin-concentrating hormone receptor 1 (MCHR-1). Synthesis and structure-activity relationships are described leading to the identification of compounds with optimized in vitro pharmacological and in vitro ADME profiles. In vivo activity has been demonstrated in animal models of food intake and depression.
Collapse
|
24
|
Guo T, Gu H, Hobbs DW, Busler DE, Rokosz LL. Discovery of tetralin ureas as potent melanin concentrating hormone 1 receptor antagonists. Bioorg Med Chem Lett 2007; 17:1718-21. [PMID: 17251014 DOI: 10.1016/j.bmcl.2006.12.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 12/14/2006] [Accepted: 12/18/2006] [Indexed: 11/23/2022]
Abstract
Melanin concentrating hormone (MCH) plays an important role in the regulation of food intake and energy balance in mammals. MCH-1 receptor (MCH1R) deficient mice are lean and resistant to diet-induced obesity. As such, MCH1R antagonists are believed to have potential as possible treatments for obesity. The discovery of a novel class of tetralin ureas as potent MCH1R antagonists is described herein.
Collapse
Affiliation(s)
- Tao Guo
- Pharmacopeia Drug Discovery Inc., PO Box 5350, Princeton, NJ 08543-5350, USA.
| | | | | | | | | |
Collapse
|
25
|
Abstract
There is compelling genetic and pharmacologic evidence to indicate that melanin-concentrating hormone receptor-1 (MCHR1) signaling is involved in the regulation of food intake and energy expenditure. The medical need for novel therapies to treat obesity and related metabolic disorders has led to a great deal of interest by pharmaceutical companies in the discovery of MCHR1 antagonists. Recent publications describing preclinical studies have demonstrated that small-molecule MCHR1 antagonists decrease food intake, bodyweight, and adiposity in rodent models of obesity. Results from ongoing early-stage clinical trials with MCHR1 antagonists are eagerly awaited, as is the movement of other MCHR1 antagonists into the clinic.
Collapse
Affiliation(s)
- Timothy J Kowalski
- Department of CV/Metabolic Diseases, Schering-Plough Research Institute, Kenilworth, New Jersey 07033, USA.
| | | |
Collapse
|
26
|
Arora S. Role of neuropeptides in appetite regulation and obesity--a review. Neuropeptides 2006; 40:375-401. [PMID: 16935329 DOI: 10.1016/j.npep.2006.07.001] [Citation(s) in RCA: 296] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 06/17/2006] [Accepted: 07/07/2006] [Indexed: 01/27/2023]
Abstract
Obesity represents the most prevalent nutritional problem worldwide which in the long run predisposes to development of diabetes mellitus, hypertension, endometrial carcinoma, osteoarthritis, gall stones and cardiovascular diseases. Despite significant reductions in dietary fat consumption, the prevalence of obesity is on a rise and is taking on pandemic proportions. Obesity develops when energy intake exceeds energy expenditure over time. Recently, a close evolutionary relationship between the peripheral and hypothalamic neuropeptides has become apparent. The hypothalamus being the central feeding organ mediates regulation of short-term and long-term dietary intake via synthesis of various orexigenic and anorectic neuropeptides. The structure and function of many hypothalamic peptides (neuropeptide Y (NPY), melanocortins, agouti-related peptide (AGRP), cocaine and amphetamine regulated transcript (CART), melanin concentrating hormone (MCH), orexins have been characterized in rodent models The peripheral neuropeptides such as cholecystokinin (CCK), ghrelin, peptide YY (PYY3-36), amylin, bombesin regulate important gastrointestinal functions such as motility, secretion, absorption, provide feedback to the central nervous system on availability of nutrients and may play a part in regulating food intake. The pharmacological potential of several endogenous peripheral peptides released prior to, during and/or after feeding are being explored. Long-term regulation is provided by the main circulating hormones leptin and insulin. These systems implicated in hypothalamic appetite regulation provide potential targets for treatment of obesity which could potentially pass into clinical development in the next 5 years. This review summarizes various effects and interrelationship of these central and peripheral neuropeptides in metabolism, obesity and their potential role as targets for treatment of obesity.
Collapse
Affiliation(s)
- Sarika Arora
- Department of Biochemistry, Lady Hardinge Medical College, Shaheed Bhagat Singh Marg, Connaught Place, New Delhi, Delhi 110 001, India.
| |
Collapse
|
27
|
Centrally Acting Anti-Obesity Agents. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2006. [DOI: 10.1016/s0065-7743(06)41005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|