1
|
Wang Y, Li D, Widjaja J, Guo R, Cai L, Yan R, Ozsoy S, Allocca G, Fang J, Dong Y, Tseng GC, Huang C, Huang YH. An Electroencephalogram Signature of Melanin-Concentrating Hormone Neuron Activities Predicts Cocaine Seeking. Biol Psychiatry 2024; 96:739-751. [PMID: 38677639 PMCID: PMC11979969 DOI: 10.1016/j.biopsych.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Identifying biomarkers that predict substance use disorder propensity may better strategize antiaddiction treatment. Melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus critically mediate interactions between sleep and substance use; however, their activities are largely obscured in surface electroencephalogram (EEG) measures, hindering the development of biomarkers. METHODS Surface EEG signals and real-time calcium (Ca2+) activities of lateral hypothalamus MCH neurons (Ca2+MCH) were simultaneously recorded in male and female adult rats. Mathematical modeling and machine learning were then applied to predict Ca2+MCH using EEG derivatives. The robustness of the predictions was tested across sex and treatment conditions. Finally, features extracted from the EEG-predicted Ca2+MCH either before or after cocaine experience were used to predict future drug-seeking behaviors. RESULTS An EEG waveform derivative-a modified theta-delta-theta peak ratio (EEGTDT ratio)-accurately tracked real-time Ca2+MCH in rats. The prediction was robust during rapid eye movement sleep (REMS), persisted through vigilance states, sleep manipulations, and circadian phases, and was consistent across sex. Moreover, cocaine self-administration and long-term withdrawal altered EEGTDT ratio, suggesting shortening and circadian redistribution of synchronous MCH neuron activities. In addition, features of EEGTDT ratio indicative of prolonged synchronous MCH neuron activities predicted lower subsequent cocaine seeking. EEGTDT ratio also exhibited advantages over conventional REMS measures for the predictions. CONCLUSIONS The identified EEGTDT ratio may serve as a noninvasive measure for assessing MCH neuron activities in vivo and evaluating REMS; it may also serve as a potential biomarker for predicting drug use propensity.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Danyang Li
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia; Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, Pennsylvania
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chengcheng Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
2
|
Li M. Is melanin-concentrating hormone in the medial preoptic area a signal for the decline of maternal care in late postpartum? Front Neuroendocrinol 2024; 75:101155. [PMID: 39222798 DOI: 10.1016/j.yfrne.2024.101155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
This manuscript proposes that melanin-concentrating hormone (MCH) in the medial preoptic area (MPOA) is an neurochemical signal evolved to trigger the declining process of maternal care. MCH in the MPOA appears only after parturition and is progressively increased with the progression of lactation, while maternal behavior declines progressively. Intra-MPOA injection of MCH decreases active maternal responses. MCH is also highly responsive to infant characteristics and maternal condition. Behavioral changes induced by MCH in late postpartum period are conducive to the decline of infant-directed maternal behavior. The MPOA MCH system may mediate the maternal behavior decline by suppressing the maternal approach motivation and/or increasing maternal withdrawal via its inhibitory action onto the mesolimbic dopamine D1/D2 receptors and its stimulating action on serotonin 5-HT2C receptors in the ventral tegmental area. Research into the MCH maternal effects will enhance our understanding of the neurochemical mechanisms underlying the maternal behavior decline.
Collapse
Affiliation(s)
- Ming Li
- Department of Psychology, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
3
|
He Q, Yuan Q, Shan H, Wu C, Gu Y, Wu K, Hu W, Zhang Y, He X, Xu HE, Zhao LH. Mechanisms of ligand recognition and activation of melanin-concentrating hormone receptors. Cell Discov 2024; 10:48. [PMID: 38710677 DOI: 10.1038/s41421-024-00679-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Melanin-concentrating hormone (MCH) is a cyclic neuropeptide that regulates food intake, energy balance, and other physiological functions by stimulating MCHR1 and MCHR2 receptors, both of which are class A G protein-coupled receptors. MCHR1 predominately couples to inhibitory G protein, Gi/o, and MCHR2 can only couple to Gq/11. Here we present cryo-electron microscopy structures of MCH-activated MCHR1 with Gi and MCH-activated MCHR2 with Gq at the global resolutions of 3.01 Å and 2.40 Å, respectively. These structures reveal that MCH adopts a consistent cysteine-mediated hairpin loop configuration when bound to both receptors. A central arginine from the LGRVY core motif between the two cysteines of MCH penetrates deeply into the transmembrane pocket, triggering receptor activation. Integrated with mutational and functional insights, our findings elucidate the molecular underpinnings of ligand recognition and MCH receptor activation and offer a structural foundation for targeted drug design.
Collapse
Affiliation(s)
- Qian He
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingning Yuan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hong Shan
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Canrong Wu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yimin Gu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Kai Wu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wen Hu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yumu Zhang
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinheng He
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Li-Hua Zhao
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
4
|
Wang Y, Li D, Widjaja J, Guo R, Cai L, Yan R, Ozsoy S, Allocca G, Fang J, Dong Y, Tseng GC, Huang C, Huang YH. An EEG Signature of MCH Neuron Activities Predicts Cocaine Seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586887. [PMID: 38586019 PMCID: PMC10996698 DOI: 10.1101/2024.03.27.586887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background Identifying biomarkers that predict substance use disorder (SUD) propensity may better strategize anti-addiction treatment. The melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus (LH) critically mediates interactions between sleep and substance use; however, their activities are largely obscured in surface electroencephalogram (EEG) measures, hindering the development of biomarkers. Methods Surface EEG signals and real-time Ca2+ activities of LH MCH neurons (Ca2+MCH) were simultaneously recorded in male and female adult rats. Mathematical modeling and machine learning were then applied to predict Ca2+MCH using EEG derivatives. The robustness of the predictions was tested across sex and treatment conditions. Finally, features extracted from the EEG-predicted Ca2+MCH either before or after cocaine experience were used to predict future drug-seeking behaviors. Results An EEG waveform derivative - a modified theta-to-delta ratio (EEG Ratio) - accurately tracks real-time Ca2+MCH in rats. The prediction was robust during rapid eye movement sleep (REMS), persisted through REMS manipulations, wakefulness, circadian phases, and was consistent across sex. Moreover, cocaine self-administration and long-term withdrawal altered EEG Ratio suggesting shortening and circadian redistribution of synchronous MCH neuron activities. In addition, features of EEG Ratio indicative of prolonged synchronous MCH neuron activities predicted lower subsequent cocaine seeking. EEG Ratio also exhibited advantages over conventional REMS measures for the predictions. Conclusions The identified EEG Ratio may serve as a non-invasive measure for assessing MCH neuron activities in vivo and evaluating REMS; it may also serve as a potential biomarker predicting drug use propensity.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Danyang Li
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | | | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia 3340
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia 3340
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia 3010
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, PA 17033
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Chengcheng Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| |
Collapse
|
5
|
Sariyer E, Sariyer AS. Computational prediction of analog compounds of the membrane protein MCHR1 antagonists ALB-127158 and KRX-104130. J Bioenerg Biomembr 2023; 55:435-446. [PMID: 37940722 DOI: 10.1007/s10863-023-09993-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/28/2023] [Indexed: 11/10/2023]
Abstract
Obesity, which is already pervasive throughout the world, endangers public health by raising the prevalence of metabolic disorders and making their treatment more difficult. The development of drugs to treat obesity is a focus of effort. Melanin concentrated hormone receptor 1 (MCHR1) is the target of some of these therapeutic possibilities since as increased levels of melanin concentrated hormone have been found in obesity models. Known MCHR1 antagonists include BMS-830216, GW-856464, NGD-4715, ALB-127158, and AMG 076, but many have failed phase-I clinical studies. As a potential treatment for cardiotoxicity, KRX-104130 has only recently been identified. As MCH system is potentially effective target for treatment of obesity, in silico research into interaction between MCHR1 and its antagonists at molecular level was the primary goal of this study. Analogues ALB-127158 and KRX-104130 were screened among the RealEnamine library. The complexes obtained by molecular docking were embedded in mimics brain-cell membrane and simulated for 540 ns, and then MM-GBSA were calculated with MMPBSA.py. With all these computational studies, similar or different aspects of selected analogous compounds to ALB-127158 and KRX-104130 were investigated. The specificity of this study was that it analyzed MCHR1 protein as embedded in membrane. It was concluded that KRX-104130's analogue Z1922310273 and ALB-127158's analogue PV-002757495233 did not cause a difference in terms of phospholipid membrane properties. In addition, all ligands remained stable in putative binding site. It has been suggested that PV-002757495233 and Z1922310273 compounds can be evaluated as MCHR1 antagonists when all these outputs are considered in melting pots.
Collapse
Affiliation(s)
- Emrah Sariyer
- Vocational School of Health Services, Medical Laboratory Techniques, Artvin Coruh University, Artvin, Turkey.
| | - Ayşegül Saral Sariyer
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Artvin Coruh University, Artvin, Turkey
| |
Collapse
|
6
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
7
|
Ruiz-Viroga V, de Ceglia M, Morelli L, Castaño EM, Calvo EB, Suárez J, Rodríguez de Fonseca F, Galeano P, Lagos P. Acute intrahippocampal administration of melanin-concentrating hormone impairs memory consolidation and decreases the expression of MCHR-1 and TrkB receptors. Prog Neuropsychopharmacol Biol Psychiatry 2023; 123:110703. [PMID: 36565982 DOI: 10.1016/j.pnpbp.2022.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Interest in the role of melanin-concentrating hormone (MCH) in memory processes has increased in recent years, with some studies reporting memory-enhancing effects, while others report deleterious effects. Due to these discrepancies, this study seeks to provide new evidence about the role of MCH in memory consolidation and its relation with BDNF/TrkB system. To this end, in the first experiment, increased doses of MCH were acutely administered in both hippocampi to groups of male rats (25, 50, 200, and 500 ng). Microinjections were carried out immediately after finishing the sample trial of two hippocampal-dependent behavioral tasks: the Novel Object Recognition Test (NORT) and the modified Elevated Plus Maze (mEPM) test. Results indicated that a dose of 200 ng of MCH or higher impaired memory consolidation in both tasks. A second experiment was performed in which a dose of 200 ng of MCH was administered alone or co-administered with the MCHR-1 antagonist ATC-0175 at the end of the sample trial in the NORT. Results showed that MCH impaired memory consolidation, while the co-administration with ATC-0175 reverted this detrimental effect. Moreover, MCH induced a significant decrease in hippocampal MCHR-1 and TrkB expression with no modification in the expression of BDNF and NMDA receptor subunits NR1, NR2A, and NR2B. These results suggest that MCH in vivo elicits pro-amnesic effects in the rat hippocampus by decreasing the availability of its receptor and TrkB receptors, thus linking both endogenous systems to memory processes.
Collapse
Affiliation(s)
- Vicente Ruiz-Viroga
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay
| | - Marialuisa de Ceglia
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo M Castaño
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo Blanco Calvo
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Campus de Teatinos S/N, Málaga 29071, Spain.
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Málaga 29071, Spain.
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Patricia Lagos
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay.
| |
Collapse
|
8
|
Vas S, Papp RS, Könczöl K, Bogáthy E, Papp N, Ádori C, Durst M, Sípos K, Ocskay K, Farkas I, Bálint F, Ferenci S, Török B, Kovács A, Szabó E, Zelena D, Kovács KJ, Földes A, Kató E, Köles L, Bagdy G, Palkovits M, Tóth ZE. Prolactin-Releasing Peptide Contributes to Stress-Related Mood Disorders and Inhibits Sleep/Mood Regulatory Melanin-Concentrating Hormone Neurons in Rats. J Neurosci 2023; 43:846-862. [PMID: 36564184 PMCID: PMC9899089 DOI: 10.1523/jneurosci.2139-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 08/31/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Stress disorders impair sleep and quality of life; however, their pathomechanisms are unknown. Prolactin-releasing peptide (PrRP) is a stress mediator; we therefore hypothesized that PrRP may be involved in the development of stress disorders. PrRP is produced by the medullary A1/A2 noradrenaline (NA) cells, which transmit stress signals to forebrain centers, and by non-NA cells in the hypothalamic dorsomedial nucleus. We found in male rats that both PrRP and PrRP-NA cells innervate melanin-concentrating hormone (MCH) producing neurons in the dorsolateral hypothalamus (DLH). These cells serve as a key hub for regulating sleep and affective states. Ex vivo, PrRP hyperpolarized MCH neurons and further increased the hyperpolarization caused by NA. Following sleep deprivation, intracerebroventricular PrRP injection reduced the number of REM sleep-active MCH cells. PrRP expression in the dorsomedial nucleus was upregulated by sleep deprivation, while downregulated by REM sleep rebound. Both in learned helplessness paradigm and after peripheral inflammation, impaired coping with sustained stress was associated with (1) overactivation of PrRP cells, (2) PrRP protein and receptor depletion in the DLH, and (3) dysregulation of MCH expression. Exposure to stress in the PrRP-insensitive period led to increased passive coping with stress. Normal PrRP signaling, therefore, seems to protect animals against stress-related disorders. PrRP signaling in the DLH is an important component of the PrRP's action, which may be mediated by MCH neurons. Moreover, PrRP receptors were downregulated in the DLH of human suicidal victims. As stress-related mental disorders are the leading cause of suicide, our findings may have particular translational relevance.SIGNIFICANCE STATEMENT Treatment resistance to monoaminergic antidepressants is a major problem. Neuropeptides that modulate the central monoaminergic signaling are promising targets for developing alternative therapeutic strategies. We found that stress-responsive prolactin-releasing peptide (PrRP) cells innervated melanin-concentrating hormone (MCH) neurons that are crucial in the regulation of sleep and mood. PrRP inhibited MCH cell activity and enhanced the inhibitory effect evoked by noradrenaline, a classic monoamine, on MCH neurons. We observed that impaired PrRP signaling led to failure in coping with chronic/repeated stress and was associated with altered MCH expression. We found alterations of the PrRP system also in suicidal human subjects. PrRP dysfunction may underlie stress disorders, and fine-tuning MCH activity by PrRP may be an important part of the mechanism.
Collapse
Affiliation(s)
- Szilvia Vas
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Semmelweis University, Budapest, 1089, Hungary
| | - Rege S Papp
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, 1094, Hungary
| | - Katalin Könczöl
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Emese Bogáthy
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
| | - Noémi Papp
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
| | - Csaba Ádori
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Máté Durst
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Klaudia Sípos
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Klementina Ocskay
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Flóra Bálint
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Szilamér Ferenci
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Bibiána Török
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Anita Kovács
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Evelin Szabó
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Dóra Zelena
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Krisztina J Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Anna Földes
- Department of Oral Biology, Semmelweis University, Budapest, 1089, Hungary
| | - Erzsébet Kató
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - László Köles
- Department of Oral Biology, Semmelweis University, Budapest, 1089, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - György Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Semmelweis University, Budapest, 1089, Hungary
- NAP2-SE New Antidepressant Target Research Group, Budapest, 1085, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, 1094, Hungary
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Zsuzsanna E Tóth
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
9
|
Guo R, Wang Y, Yan R, Chen B, Ding W, Gorczyca MT, Ozsoy S, Cai L, Hines RL, Tseng GC, Allocca G, Dong Y, Fang J, Huang YH. Rapid Eye Movement Sleep Engages Melanin-Concentrating Hormone Neurons to Reduce Cocaine Seeking. Biol Psychiatry 2022; 92:880-894. [PMID: 35953320 PMCID: PMC9872495 DOI: 10.1016/j.biopsych.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Persistent sleep disruptions following withdrawal from abused drugs may hold keys to battle drug relapse. It is posited that there may be sleep signatures that predict relapse propensity, identifying which may open new avenues for treating substance use disorders. METHODS We trained male rats (approximately postnatal day 56) to self-administer cocaine. After long-term drug withdrawal (approximately postnatal day 100), we examined the correlations between the intensity of cocaine seeking and key sleep features. To test for causal relationships, we then used behavioral, chemogenetic, or optogenetic methods to selectively increase rapid eye movement sleep (REMS) and measured behavioral and electrophysiological outcomes to probe for cellular and circuit mechanisms underlying REMS-mediated regulation of cocaine seeking. RESULTS A selective set of REMS features was preferentially associated with the intensity of cue-induced cocaine seeking after drug withdrawal. Moreover, selectively increasing REMS time and continuity by environmental warming attenuated a withdrawal time-dependent intensification of cocaine seeking, or incubation of cocaine craving, suggesting that REMS may benefit withdrawal. Warming increased the activity of lateral hypothalamic melanin-concentrating hormone (MCH) neurons selectively during prolonged REMS episodes and counteracted cocaine-induced synaptic accumulation of calcium-permeable AMPA receptors in the nucleus accumbens-a critical substrate for incubation. Finally, the warming effects were partly mimicked by chemogenetic or optogenetic stimulations of MCH neurons during sleep, or intra-accumbens infusions of MCH peptide during the rat's inactive phase. CONCLUSIONS REMS may encode individual vulnerability to relapse, and MCH neuron activities can be selectively targeted during REMS to reduce drug relapse.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bo Chen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wanqiao Ding
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael T Gorczyca
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel L Hines
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia; Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, Pennsylvania
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
10
|
Kupcova I, Danisovic L, Grgac I, Harsanyi S. Anxiety and Depression: What Do We Know of Neuropeptides? Behav Sci (Basel) 2022; 12:262. [PMID: 36004833 PMCID: PMC9405013 DOI: 10.3390/bs12080262] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
In modern society, there has been a rising trend of depression and anxiety. This trend heavily impacts the population's mental health and thus contributes significantly to morbidity and, in the worst case, to suicides. Modern medicine, with many antidepressants and anxiolytics at hand, is still unable to achieve remission in many patients. The pathophysiology of depression and anxiety is still only marginally understood, which encouraged researchers to focus on neuropeptides, as they are a vast group of signaling molecules in the nervous system. Neuropeptides are involved in the regulation of many physiological functions. Some act as neuromodulators and are often co-released with neurotransmitters that allow for reciprocal communication between the brain and the body. Most studied in the past were the antidepressant and anxiolytic effects of oxytocin, vasopressin or neuropeptide Y and S, or Substance P. However, in recent years, more and more novel neuropeptides have been added to the list, with implications for the research and development of new targets, diagnostic elements, and even therapies to treat anxiety and depressive disorders. In this review, we take a close look at all currently studied neuropeptides, their related pathways, their roles in stress adaptation, and the etiology of anxiety and depression in humans and animal models. We will focus on the latest research and information regarding these associated neuropeptides and thus picture their potential uses in the future.
Collapse
Affiliation(s)
- Ida Kupcova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| | - Ivan Grgac
- Institute of Anatomy, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| |
Collapse
|
11
|
Kato Y, Katsumata H, Inutsuka A, Yamanaka A, Onaka T, Minami S, Orikasa C. Involvement of MCH-oxytocin neural relay within the hypothalamus in murine nursing behavior. Sci Rep 2021; 11:3348. [PMID: 33558633 PMCID: PMC7870840 DOI: 10.1038/s41598-021-82773-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Multiple sequential actions, performed during parental behaviors, are essential elements of reproduction in mammalian species. We showed that neurons expressing melanin concentrating hormone (MCH) in the lateral hypothalamic area (LHA) are more active in rodents of both sexes when exhibiting parental nursing behavior. Genetic ablation of the LHA-MCH neurons impaired maternal nursing. The post-birth survival rate was lower in pups born to female mice with congenitally ablated MCH neurons under control of tet-off system, exhibiting reduced crouching behavior. Virgin female and male mice with ablated MCH neurons were less interested in pups and maternal care. Chemogenetic and optogenetic stimulation of LHA-MCH neurons induced parental nursing in virgin female and male mice. LHA-MCH GABAergic neurons project fibres to the paraventricular hypothalamic nucleus (PVN) neurons. Optogenetic stimulation of PVN induces nursing crouching behavior along with increasing plasma oxytocin levels. The hypothalamic MCH neural relays play important functional roles in parental nursing behavior in female and male mice.
Collapse
Affiliation(s)
- Yoko Kato
- Department of Bioregulation, Institute for Advanced Medical Science, Nippon Medical School, Kawasaki, 211-8533, Japan
| | - Harumi Katsumata
- Department of Bioregulation, Institute for Advanced Medical Science, Nippon Medical School, Kawasaki, 211-8533, Japan
| | - Ayumu Inutsuka
- Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan
| | - Tatsushi Onaka
- Department of Physiology, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shiro Minami
- Department of Bioregulation, Institute for Advanced Medical Science, Nippon Medical School, Kawasaki, 211-8533, Japan
| | - Chitose Orikasa
- Department of Bioregulation, Institute for Advanced Medical Science, Nippon Medical School, Kawasaki, 211-8533, Japan.
| |
Collapse
|
12
|
Sharma R, Sharma A, Sahota P, Thakkar MM. Orexin gene expression is downregulated in alcohol dependent rats during acute alcohol withdrawal. Neurosci Lett 2020; 739:135347. [PMID: 33011195 DOI: 10.1016/j.neulet.2020.135347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 01/14/2023]
Abstract
Alcohol use disorders (AUD) are chronic relapsing brain disorder characterized by compulsive and heavy alcohol consumption. During acute withdrawal, patients with AUD display excessive daytime sleepiness, a condition linked to serious life-threatening complications, however, the mechanism is not known. Orexin and melanin-concentrating hormone (MCH) are the two hypothalamic neuropeptides that regulate many behaviors including sleep-wakefulness, and alcohol consumption, reinforcement, and reinstatement. Importantly, loss of orexin neurons causes narcolepsy, a severe sleep disorder with excessive daytime sleepiness. Does acute alcohol withdrawal reduce orexin gene expression? To investigate this, male Sprague-Dawley rats were divided in two groups: Rats were either administered with alcohol, mixed with infant formula (alcohol group) or control mixture containing water and infant formula (Controls) by gastric intubation every 8 h for 4 days using Majchrowicz's chronic binge drinking protocol. The doses of alcohol were adjusted depending on degree of intoxication, exhibited by animals, prior to each dose. The animals were euthanized after 12 h of last alcohol/water administration. During withdrawal, the hypothalamus was rapidly dissected out, and the expressions of orexin and MCH genes were examined by Real-time PCR. There was a significant reduction in orexin gene expression in rats subjected to alcohol withdrawal as compared to controls. No such change was observed in the MCH gene expression. These results suggest that downregulation of orexin gene expression may be a possible mechanism responsible for excessive daytime sleepiness associated with alcohol withdrawal in patients with AUD.
Collapse
Affiliation(s)
- Rishi Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, 65201, United States
| | - Abhilasha Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, 65201, United States
| | - Pradeep Sahota
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, 65201, United States
| | - Mahesh M Thakkar
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, 65201, United States.
| |
Collapse
|
13
|
Individual Mesopontine Neurons Implicated in Anesthetic Loss-of-consciousness Employ Separate Ascending Pathways to the Cerebral Cortex. Neuroscience 2020; 432:188-204. [DOI: 10.1016/j.neuroscience.2020.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 01/19/2023]
|
14
|
Cengiz M, Karaj V, Kocabasoğlu N, Gozubatik-Celik G, Dirican A, Bayoglu B. Orexin/hypocretin receptor, Orx1, gene variants are associated with major depressive disorder. Int J Psychiatry Clin Pract 2019; 23:114-121. [PMID: 30596528 DOI: 10.1080/13651501.2018.1551549] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: Orexins (hypocretins) are neuropeptides expressed in hypothalamic neurons and have regulatory roles in feeding/drinking behaviours, endocrine functions and sleep/wakefulness state. Major depressive disorder (MDD) is a major mood disorder and neurotransmitter dysfunction in hypothalamic neurons may have roles in its formation. Hence, we conducted experiments to determine whether orexin receptor 1 and 2 (Orx1, Orx2) genes were associated with MDD development. Methods: Seventy-five MDD patients and 87 healthy controls were enrolled for the study. Genotyping was carried out with real-time polymerase chain reaction (RT-PCR). Hamilton Rating-Scale for Depression (HRSD) and Beck Depression Inventory (BDI) were utilized to evaluate depressive symptom severity. Results: A significant relation was found in genotype frequencies of Orx1 rs10914456 and rs2271933 variants between MDD patients and controls (p = .009, p = .006). Rs10914456 CC genotype increased MDD risk 3.57 times more than carrying other genotypes (p = .008, OR =3.57;95% CI: 1.39-9.14). However, no association was observed in Orx2 rs2653349 genotypes for MDD development (p > .05). Although statistically not significant, HRSD scores were diminished in MDD subjects carrying rs10914456 CC variants when compared with CT and TT variants (p = .069). Conclusion. This study suggests that, Orx1 rs10914456 and rs2271933 can be associated with MDD development. Hence, Orx1 rs10914456 variants may affect depressive symptom severity.
Collapse
Affiliation(s)
- Mujgan Cengiz
- a Department of Medical Biology, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Vilson Karaj
- b Department of Science, Institute of Forensic Sciences , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Nese Kocabasoğlu
- c Department of Psychiatry, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Gokcen Gozubatik-Celik
- d Department of Neurology, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Ahmet Dirican
- e Department of Biostatistics and Medical Informatics, Istanbul Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Burcu Bayoglu
- a Department of Medical Biology, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| |
Collapse
|
15
|
Balkan B, Pogun S. Nicotinic Cholinergic System in the Hypothalamus Modulates the Activity of the Hypothalamic Neuropeptides During the Stress Response. Curr Neuropharmacol 2018; 16:371-387. [PMID: 28730966 PMCID: PMC6018196 DOI: 10.2174/1570159x15666170720092442] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The hypothalamus harbors high levels of cholinergic neurons and axon terminals. Nicotinic acetylcholine receptors, which play an important role in cholinergic neurotransmission, are expressed abundantly in the hypothalamus. Accumulating evidence reveals a regulatory role for nicotine in the regulation of the stress responses. The present review will discuss the hypothalamic neuropeptides and their interaction with the nicotinic cholinergic system. The anatomical distribution of the cholinergic neurons, axon terminals and nicotinic receptors in discrete hypothalamic nuclei will be described. The effect of nicotinic cholinergic neurotransmission and nicotine exposure on hypothalamic-pituitaryadrenal (HPA) axis regulation at the hypothalamic level will be analyzed in view of the different neuropeptides involved. METHODS Published research related to nicotinic cholinergic regulation of the HPA axis activity at the hypothalamic level is reviewed. RESULTS The nicotinic cholinergic system is one of the major modulators of the HPA axis activity. There is substantial evidence supporting the regulation of hypothalamic neuropeptides by nicotinic acetylcholine receptors. However, most of the studies showing the nicotinic regulation of hypothalamic neuropeptides have employed systemic administration of nicotine. Additionally, we know little about the nicotinic receptor distribution on neuropeptide-synthesizing neurons in the hypothalamus and the physiological responses they trigger in these neurons. CONCLUSION Disturbed functioning of the HPA axis and hypothalamic neuropeptides results in pathologies such as depression, anxiety disorders and obesity, which are common and significant health problems. A better understanding of the nicotinic regulation of hypothalamic neuropeptides will aid in drug development and provide means to cope with these diseases. Considering that nicotine is also an abused substance, a better understanding of the role of the nicotinic cholinergic system on the HPA axis will aid in developing improved therapeutic strategies for smoking cessation.
Collapse
Affiliation(s)
- Burcu Balkan
- Center for Brain Research, Ege University, Bornova, Izmir, Turkey.,Department of Physiology, School of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Sakire Pogun
- Center for Brain Research, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
16
|
Bencze J, Pocsai K, Murnyák B, Gergely PA, Juhász B, Szilvássy Z, Hortobágyi T. The Melanin-concentrating Hormone System in Human, Rodent and Avian Brain. Open Med (Wars) 2018; 13:264-269. [PMID: 29992190 PMCID: PMC6034100 DOI: 10.1515/med-2018-0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/06/2018] [Indexed: 12/20/2022] Open
Abstract
Melanin-concentrating hormone (MCH) is a cyclic 19 amino acid orexigenic hypothalamic peptide. MCH is located in the lateral and dorsal hypothalamus, as well as in the zona incerta. In mammals MCH increases food intake, contributes to regulation of energy balance, temperature, reproductive function, endocrine homeostasis and biological rhythms. Several studies have proved the significance of MCH in obesity, diabetes and depression. Although the peptide is well-characterized in mouse models, much less is known about its functions in avians. In birds the MCH system especially in the lateral and basal hypothalamus has important connections to the limbic system and it coordinates the vegetative and endocrine functions, as well as the emotional behaviour. Pharmacological modulation of MCH system could contribute to the therapy of eating disorders and improve agricultural efficiency regarding avians. Reviewing the current knowledge on MCH system in human, rodents and avians may stimulate a new wave of studies in the field.
Collapse
Affiliation(s)
- János Bencze
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Krisztina Pocsai
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Murnyák
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Debrecen, Hungary.,Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Attila Gergely
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Tibor Hortobágyi
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Nagyerdei krt. 98., H-4032, Hungary.,Department of Pathology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Old Age Psychiatry, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, UK.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Debrecen, Hungary
| |
Collapse
|
17
|
Li W, Papilloud A, Lozano-Montes L, Zhao N, Ye X, Zhang X, Sandi C, Rainer G. Stress Impacts the Regulation Neuropeptides in the Rat Hippocampus and Prefrontal Cortex. Proteomics 2018; 18:e1700408. [PMID: 29406625 DOI: 10.1002/pmic.201700408] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/10/2018] [Indexed: 11/05/2022]
Abstract
Adverse life experiences increase the lifetime risk to several stress-related psychopathologies, such as anxiety or depressive-like symptoms following stress in adulthood. However, the neurochemical modulations triggered by stress have not been fully characterized. Neuropeptides play an important role as signaling molecules that contribute to physiological regulation and have been linked to neurological and psychiatric diseases. However, little is known about the influence of stress on neuropeptide regulation in the brain. Here, we have performed an exploratory study of how neuropeptide expression at adulthood is modulated by experiencing a period of multiple stressful experiences. We have targeted hippocampus and prefrontal cortex (PFC) brain areas, which have previously been shown to be modulated by stressors, employing a targeted liquid chromatography-mass spectrometry (LC-MS) based approach that permits broad peptide coverage with high sensitivity. We found that in the hippocampus, Met-enkephalin, Met-enkephalin-Arg-Phe, and Met-enkephalin-Arg-Gly-Leu were upregulated, while Leu-enkephalin and Little SAAS were downregulated after stress. In the PFC area, Met-enkephalin-Arg-Phe, Met-enkephalin-Arg-Gly-Leu, peptide PHI-27, somatostatin-28 (AA1-12), and Little SAAS were all downregulated. This systematic evaluation of neuropeptide alterations in the hippocampus and PFC suggests that stressors impact neuropeptides and that neuropeptide regulation is brain-area specific. These findings suggest several potential peptide candidates, which warrant further investigations in terms of correlation with depression-associated behaviors.
Collapse
Affiliation(s)
- Wenxue Li
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Aurelie Papilloud
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Science, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | | | - Nan Zhao
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Xueting Ye
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Xiaozhe Zhang
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Science, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Gregor Rainer
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
18
|
Sanathara NM, Garau C, Alachkar A, Wang L, Wang Z, Nishimori K, Xu X, Civelli O. Melanin concentrating hormone modulates oxytocin-mediated marble burying. Neuropharmacology 2018; 128:22-32. [PMID: 28888943 PMCID: PMC5830107 DOI: 10.1016/j.neuropharm.2017.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/15/2017] [Accepted: 09/05/2017] [Indexed: 11/27/2022]
Abstract
Repetitive and perseverative behaviors are common features of a number of neuropsychiatric diseases such as Angelman's syndrome, Tourette's syndrome, obsessive-compulsive disorder, and autism spectrum disorders. The oxytocin system has been linked to the regulation of repetitive behavior in both animal models and humans, but many of its downstream targets have still to be found. We report that the melanin-concentrating hormone (MCH) system is a target of the oxytocin system in regulating one repetitive behavior, marble burying. First we report that nearly 60% of MCH neurons express oxytocin receptors, and demonstrate using rabies mediated tract tracing that MCH neurons receive direct presynaptic input from oxytocin neurons. Then we show that MCH receptor knockout (MCHR1KO) mice and MCH ablated animals display increased marble burying response while central MCH infusion decreases it. Finally, we demonstrate the downstream role of the MCH system on oxytocin mediated marble burying by showing that central infusions of MCH and oxytocin alone or together reduce it while antagonizing the MCH system blocks oxytocin-mediated reduction of this behavior. Our findings reveal a novel role for the MCH system as a mediator of the role of oxytocin in regulating marble-burying behavior in mice.
Collapse
Affiliation(s)
- Nayna M Sanathara
- Department of Pharmacology, University of California, Irvine, CA, 92697, USA
| | - Celia Garau
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Amal Alachkar
- Department of Pharmacology, University of California, Irvine, CA, 92697, USA
| | - Lien Wang
- Department of Pharmacology, University of California, Irvine, CA, 92697, USA
| | - Zhiwei Wang
- Department of Pharmacology, University of California, Irvine, CA, 92697, USA
| | - Katsuhiko Nishimori
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, Sendai, Miyagi, 981-8555, Japan
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Olivier Civelli
- Department of Pharmacology, University of California, Irvine, CA, 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA; Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
19
|
Luan X, Sun X, Guo F, Zhang D, Wang C, Ma L, Xu L. Lateral hypothalamic Orexin-A-ergic projections to the arcuate nucleus modulate gastric functionin vivo. J Neurochem 2017; 143:697-707. [PMID: 28984906 DOI: 10.1111/jnc.14233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/18/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Xiao Luan
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Xiangrong Sun
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Feifei Guo
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Di Zhang
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Cheng Wang
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Li Ma
- Departmemt of Clinical Nutrition; Affiliated Hospital; Qingdao University; Qingdao China
| | - Luo Xu
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| |
Collapse
|
20
|
Xie Y, Kaufmann D, Moulton MJ, Panahi S, Gaynes JA, Watters HN, Zhou D, Xue HH, Fung CM, Levine EM, Letsou A, Brennan KC, Dorsky RI. Lef1-dependent hypothalamic neurogenesis inhibits anxiety. PLoS Biol 2017; 15:e2002257. [PMID: 28837622 PMCID: PMC5570277 DOI: 10.1371/journal.pbio.2002257] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/21/2017] [Indexed: 11/19/2022] Open
Abstract
While innate behaviors are conserved throughout the animal kingdom, it is unknown whether common signaling pathways regulate the development of neuronal populations mediating these behaviors in diverse organisms. Here, we demonstrate that the Wnt/ß-catenin effector Lef1 is required for the differentiation of anxiolytic hypothalamic neurons in zebrafish and mice, although the identity of Lef1-dependent genes and neurons differ between these 2 species. We further show that zebrafish and Drosophila have common Lef1-dependent gene expression in their respective neuroendocrine organs, consistent with a conserved pathway that has diverged in the mouse. Finally, orthologs of Lef1-dependent genes from both zebrafish and mouse show highly correlated hypothalamic expression in marmosets and humans, suggesting co-regulation of 2 parallel anxiolytic pathways in primates. These findings demonstrate that during evolution, a transcription factor can act through multiple mechanisms to generate a common behavioral output, and that Lef1 regulates circuit development that is fundamentally important for mediating anxiety in a wide variety of animal species.
Collapse
Affiliation(s)
- Yuanyuan Xie
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Dan Kaufmann
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Matthew J. Moulton
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
| | - Samin Panahi
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - John A. Gaynes
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Harrison N. Watters
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Dingxi Zhou
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
- School of Life Sciences, Peking University, Beijing, China
| | - Hai-Hui Xue
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Camille M. Fung
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Edward M. Levine
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States of America
| | - Anthea Letsou
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
| | - K. C. Brennan
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Richard I. Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
21
|
Xu L, Wang H, Gong Y, Pang M, Sun X, Guo F, Gao S. Nesfatin-1 regulates the lateral hypothalamic area melanin-concentrating hormone-responsive gastric distension-sensitive neurons and gastric function via arcuate nucleus innervation. Metabolism 2017; 67:14-25. [PMID: 28081774 DOI: 10.1016/j.metabol.2016.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/18/2016] [Accepted: 10/23/2016] [Indexed: 12/15/2022]
Abstract
Nesfatin-1, a recently discovered neuropeptide involved in satiety. Recent studies have revealed that central nesfatin-1 inhibits gastric emptying and gastric acid secretion, though the mechanisms involved in these processes are not known. We aim to explore the effects of nesfatin-1 on a population of gastric distension (GD)-sensitive neurons in the lateral hypothalamus (LHA), gastric motility, and gastric secretion and the role for an arcuate nucleus (Arc)-LHA neural pathway in these processes. Single unit extracellular discharge recordings were made in of LHA. Further, gastric motility and gastric secretion in rats were monitored. Retrograde tracing and fluorescent immunohistochemical staining were used to explore nesfatin-1 neuron projection. The results revealed that administration of nesfatin-1 to the LHA or electric stimulation of the Arc could alter the neuronal activity of melanin-concentrating hormone (MCH)-responsive, GD-responsive neurons in LHA, which could be blocked by pretreatment with MCH receptor-1 antagonist PMC-3881-PI or weakened by pretreatment of a nesfatin-1 antibody in LHA. Administration of nesfatin-1 into LHA could inhibit gastric motility and gastric secretion, and these effects could be enhanced by administration of PMC-3881-PI. Electrical stimulation of Arc promoted the gastric motility and gastric secretion. Nesfatin-1 antibody or PMC-3881-PI pretreatment to LHA had no effect on Arc stimulation-induced gastric motility, but these pretreatments did alter Arc stimulation-induced effects on gastric secretion. Our findings suggest that nesfatin-1 signaling in LHA participates in the regulation of efferent information from the gastrointestinal tract and gastric secretion which also involve MCH signaling. Further, they show that a nesfatin-1-positive Arc to LHA pathway is critical for these effects.
Collapse
Affiliation(s)
- Luo Xu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, Shandong, China.
| | - Hongbo Wang
- Department of Gastroenterology, Jimo People's Hospital, Qingdao, Shandong, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, Shandong, China
| | - Mingjie Pang
- Department of Otolaryngology, Qingdao Municipal Hospital (Group), Qingdao, Shandong, China
| | - Xiangrong Sun
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Feifei Guo
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Shengli Gao
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
22
|
Eiler WJA, Chen Y, Slieker LJ, Ardayfio PA, Statnick MA, Witkin JM. Consequences of constitutive deletion of melanin-concentrating hormone-1 receptors for feeding and foraging behaviors of mice. Behav Brain Res 2016; 316:271-278. [PMID: 27633558 DOI: 10.1016/j.bbr.2016.09.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/04/2016] [Accepted: 09/11/2016] [Indexed: 01/01/2023]
Abstract
In order to decipher the functional involvement of melanin-concentrating hormone 1 (MCH1) receptors in the control of feeding and foraging behaviors, mice with constitutive deletion of MCH1 receptors MCH1R -/- or knockout (KO) were studied and compared to age-matched littermate control mice (MCH1R +/+ or wildtype (WT)). Several challenges to food-motivated behaviors of food-restricted WT and KO mice were implemented. There were no differences between genotypes in the acquisition of a nose-poke response that produced food or in a discrimination between a response that produced food and one that did not. There were also no genotype differences in the rate of extinction of a food-motivated response. However, during the first day of extinction, foraging behaviors were increased significantly more in KO than in WT mice. Likewise, when the response requirement to obtain food was progressively increased, KO mice made significantly more food-directed responses than WT mice. Although adulteration of food with quinine did not suppress food-directed behavior in either genotype when the mice were food-restricted, manipulation of the degree of food-deprivation resulted in suppression of behavior of WT mice without suppressing the behavior of KO mice. Although response-produced foot shock suppressed food-maintained responding of both WT and KO mice, equipotent levels of shock (based upon psychophysical thresholds) suppressed behavior of WT mice without suppressing behavior of the KO mice. Finally, under a Vogel conflict procedure, KO mice had significantly higher levels of both punished and non-punished food maintained responding. Thus, the data from challenges with both appetitive and noxious stimulus challenges support the conclusion that mice with constitutive deletion of MCH1Rs have increased food seeking motivation that is coincident with their higher metabolism. The data also highlight important differences in the biological impact of MCH1 receptor KO and MCH1 receptor antagonism.
Collapse
Affiliation(s)
- William J A Eiler
- Divisions of Neuroscience, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, United States
| | - Yanyun Chen
- Endocrinology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, United States
| | - Lawrence J Slieker
- Endocrinology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, United States
| | - Paul A Ardayfio
- Divisions of Neuroscience, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, United States
| | - Michael A Statnick
- Endocrinology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, United States
| | - Jeffrey M Witkin
- Divisions of Neuroscience, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, United States.
| |
Collapse
|
23
|
Evolution of physicochemical properties of melanin concentrating hormone receptor 1 (MCHr1) antagonists. Bioorg Med Chem Lett 2016; 26:4559-4564. [PMID: 27595423 DOI: 10.1016/j.bmcl.2016.08.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/12/2016] [Accepted: 08/20/2016] [Indexed: 02/07/2023]
Abstract
One pharmacological principle for the treatment of obesity is blockade of the melanin concentrating hormone receptor 1 (MCHr1), which in rodents has been shown to be strongly associated with food intake and energy expenditure. However, discovery of safe and efficacious MCHr1 antagonists has proved to be complex. So far, six compounds have been progressed into clinical trials, but clinical validation of the concept is still lacking. An account of discovery of the three most recent clinical candidates targeting the MCHr1 receptor is given, with an emphasis on their physicochemical properties.
Collapse
|
24
|
Shoda T, Futamura K, Orihara K, Emi-Sugie M, Saito H, Matsumoto K, Matsuda A. Recent advances in understanding the roles of vascular endothelial cells in allergic inflammation. Allergol Int 2016; 65:21-9. [PMID: 26666487 DOI: 10.1016/j.alit.2015.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/30/2015] [Accepted: 08/10/2015] [Indexed: 12/14/2022] Open
Abstract
Allergic disorders commonly involve both chronic tissue inflammation and remodeling caused by immunological reactions to various antigens on tissue surfaces. Due to their anatomical location, vascular endothelial cells are the final responders to interact with various exogenous factors that come into contact with the epithelial surface, such as pathogen-associated molecular patterns (PAMPs) and antigens. Recent studies have shed light on the important roles of endothelial cells in the development and exacerbation of allergic disorders. For instance, endothelial cells have the greatest potential to produce several key molecules that are deeply involved in allergic inflammation, such as periostin and thymus and activation-regulated chemokine (TARC/CCL17). Additionally, endothelial cells were recently shown to be important functional targets for IL-33--an essential regulator of allergic inflammation. Notably, almost all endothelial cell responses and functions involved in allergic inflammation are not suppressed by corticosteroids. These corticosteroid-refractory endothelial cell responses and functions include TNF-α-associated angiogenesis, leukocyte adhesion, IL-33-mediated responses and periostin and TARC production. Therefore, these unique responses and functions of endothelial cells may be critically involved in the pathogenesis of various allergic disorders, especially their refractory processes. Here, we review recent studies, including ours, which have elucidated previously unknown pathophysiological roles of vascular endothelial cells in allergic inflammation and discuss the possibility of endothelium-targeted therapy for allergic disorders.
Collapse
Affiliation(s)
- Tetsuo Shoda
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Kyoko Futamura
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kanami Orihara
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Maiko Emi-Sugie
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hirohisa Saito
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Akio Matsuda
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
25
|
Torterolo P, Scorza C, Lagos P, Urbanavicius J, Benedetto L, Pascovich C, López-Hill X, Chase MH, Monti JM. Melanin-Concentrating Hormone (MCH): Role in REM Sleep and Depression. Front Neurosci 2015; 9:475. [PMID: 26733789 PMCID: PMC4681773 DOI: 10.3389/fnins.2015.00475] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/26/2015] [Indexed: 12/05/2022] Open
Abstract
The melanin-concentrating hormone (MCH) is a peptidergic neuromodulator synthesized by neurons of the lateral sector of the posterior hypothalamus and zona incerta. MCHergic neurons project throughout the central nervous system, including areas such as the dorsal (DR) and median (MR) raphe nuclei, which are involved in the control of sleep and mood. Major Depression (MD) is a prevalent psychiatric disease diagnosed on the basis of symptomatic criteria such as sadness or melancholia, guilt, irritability, and anhedonia. A short REM sleep latency (i.e., the interval between sleep onset and the first REM sleep period), as well as an increase in the duration of REM sleep and the density of rapid-eye movements during this state, are considered important biological markers of depression. The fact that the greatest firing rate of MCHergic neurons occurs during REM sleep and that optogenetic stimulation of these neurons induces sleep, tends to indicate that MCH plays a critical role in the generation and maintenance of sleep, especially REM sleep. In addition, the acute microinjection of MCH into the DR promotes REM sleep, while immunoneutralization of this peptide within the DR decreases the time spent in this state. Moreover, microinjections of MCH into either the DR or MR promote a depressive-like behavior. In the DR, this effect is prevented by the systemic administration of antidepressant drugs (either fluoxetine or nortriptyline) and blocked by the intra-DR microinjection of a specific MCH receptor antagonist. Using electrophysiological and microdialysis techniques we demonstrated also that MCH decreases the activity of serotonergic DR neurons. Therefore, there are substantive experimental data suggesting that the MCHergic system plays a role in the control of REM sleep and, in addition, in the pathophysiology of depression. Consequently, in the present report, we summarize and evaluate the current data and hypotheses related to the role of MCH in REM sleep and MD.
Collapse
Affiliation(s)
- Pablo Torterolo
- Department of Physiology, School of Medicine, Universidad de la República Montevideo, Uruguay
| | - Cecilia Scorza
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Patricia Lagos
- Department of Physiology, School of Medicine, Universidad de la República Montevideo, Uruguay
| | - Jessika Urbanavicius
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Luciana Benedetto
- Department of Physiology, School of Medicine, Universidad de la República Montevideo, Uruguay
| | - Claudia Pascovich
- Department of Physiology, School of Medicine, Universidad de la República Montevideo, Uruguay
| | - Ximena López-Hill
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Michael H Chase
- WebSciences International and University of California, Los Angeles School of Medicine Los Angeles, CA, USA
| | - Jaime M Monti
- Department of Pharmacology and Therapeutics, School of Medicine, Hospital de Clínicas, Universidad de la República Montevideo, Uruguay
| |
Collapse
|
26
|
Barandas R, Landgraf D, McCarthy MJ, Welsh DK. Circadian Clocks as Modulators of Metabolic Comorbidity in Psychiatric Disorders. Curr Psychiatry Rep 2015; 17:98. [PMID: 26483181 DOI: 10.1007/s11920-015-0637-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Psychiatric disorders such as schizophrenia, bipolar disorder, and major depressive disorder are often accompanied by metabolic dysfunction symptoms, including obesity and diabetes. Since the circadian system controls important brain systems that regulate affective, cognitive, and metabolic functions, and neuropsychiatric and metabolic diseases are often correlated with disturbances of circadian rhythms, we hypothesize that dysregulation of circadian clocks plays a central role in metabolic comorbidity in psychiatric disorders. In this review paper, we highlight the role of circadian clocks in glucocorticoid, dopamine, and orexin/melanin-concentrating hormone systems and describe how a dysfunction of these clocks may contribute to the simultaneous development of psychiatric and metabolic symptoms.
Collapse
Affiliation(s)
- Rita Barandas
- Department of Psychiatry, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA
| | - Dominic Landgraf
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA.
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA.
| | - Michael J McCarthy
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA
| | - David K Welsh
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA
| |
Collapse
|
27
|
Feltelius N, Persson I, Ahlqvist-Rastad J, Andersson M, Arnheim-Dahlström L, Bergman P, Granath F, Adori C, Hökfelt T, Kühlmann-Berenzon S, Liljeström P, Maeurer M, Olsson T, Örtqvist Å, Partinen M, Salmonson T, Zethelius B. A coordinated cross-disciplinary research initiative to address an increased incidence of narcolepsy following the 2009-2010 Pandemrix vaccination programme in Sweden. J Intern Med 2015; 278:335-53. [PMID: 26123389 DOI: 10.1111/joim.12391] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In response to the 2009-2010 influenza A(H1N1)pdm09 pandemic, a mass vaccination programme with the AS03-adjuvanted influenza A(H1N1) vaccine Pandemrix was initiated in Sweden. Unexpectedly, there were a number of narcolepsy cases amongst vaccinated children and adolescents reported. In this review, we summarize the results of a joint cross-disciplinary national research effort to investigate the adverse reaction signal from the spontaneous reporting system and to better understand possible causative mechanisms. A three- to fourfold increased risk of narcolepsy in vaccinated children and adolescents was verified by epidemiological studies. Of importance, no risk increase was observed for the other neurological and autoimmune diseases studied. Genetic studies confirmed the association with the allele HLA-DQB1*06:02, which is known to be related to sporadic narcolepsy. Furthermore, a number of studies using cellular and molecular experimental models investigated possible links between influenza vaccination and narcolepsy. Serum analysis, using a peptide microarray platform, showed that individuals who received Pandemrix exhibited a different epitope reactivity pattern to neuraminidase and haemagglutinin, as compared to individuals who were infected with H1N1. Patients with narcolepsy were also found to have increased levels of interferon-gamma production in response to streptococcus-associated antigens. The chain of patient-related events and the study results emerging over time were subjected to intense nationwide media attention. The importance of transparent communication and collaboration with patient representatives to maintain public trust in vaccination programmes is also discussed in the review. Organizational challenges due to this unexpected event delayed the initiation of some of the research projects, still the main objectives of this joint, cross-disciplinary research effort were reached, and important insights were acquired for future, similar situations in which a fast and effective task force may be required to evaluate vaccination-related adverse events.
Collapse
Affiliation(s)
| | - I Persson
- Medical Products Agency, Uppsala, Sweden
| | | | | | - L Arnheim-Dahlström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - P Bergman
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - F Granath
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - C Adori
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - T Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - P Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - M Maeurer
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - T Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Å Örtqvist
- Department of Communicable Disease Control and Prevention, Stockholm County Council, Stockholm, Sweden.,Department of Medicine, Unit of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - M Partinen
- Helsinki Sleep Clinic, Vitalmed Research Centre, Helsinki, Finland.,Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
28
|
Müller SG, Heckel A, Kley JT, Lehmann T, Lustenberger P, Oost T, Roth GJ, Rudolf K, Arndt K, Lenter M, Lotz RR, Maier GM, Markert M, Schindler M, Stenkamp D. Design, synthesis and evaluation of MCH receptor 1 antagonists—Part I: Optimization of HTS hits towards an in vivo efficacious tool compound BI 414. Bioorg Med Chem Lett 2015; 25:3264-9. [DOI: 10.1016/j.bmcl.2015.05.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 12/14/2022]
|
29
|
Schmidt FM, Nowak C, Kratzsch J, Sander C, Hegerl U, Schönknecht P. Dynamics of melanin-concentrating hormone (MCH) serum levels in major depressive disorder during antidepressant treatment. J Affect Disord 2015; 180:207-13. [PMID: 25932975 DOI: 10.1016/j.jad.2015.03.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 03/22/2015] [Accepted: 03/24/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND In preclinical studies, the hypothalamic polypeptide melanin-concentrating hormone (MCH) has been shown to be involved in depression-like behavior and modulations of MCH and MCH-receptors were proposed as potential new antidepressant drug targets. METHODS For the first time, MCH serum levels were explored in 30 patients with major depressive disorder (MDD) prior to (T1) and after 2 (T2) and 4 weeks (T3) of antidepressant treatment and in 30 age- and sex-matched healthy controls by applying a fluorescence immunoassay. RESULTS Levels of MCH did not differ significantly between un-medicated patients (444.11±174.63pg/mL SD) and controls (450.68±210.03pg/mL SD). In MDD patients, MCH levels significantly decreased from T1 to T3 (F=4.663; p=0.013). Post-hoc analyses showed that these changes were limited to patients treated with mirtazapine but not escitalopram and female but not male patients. MCH-levels showed high correlations from T1 to T3 (r≥0.964, p<0.001) and were found to correlate significantly with parameters of sleep within the controls. LIMITATIONS Small sample size. No follow-up measures were performed within the control group. CONCLUSIONS Our findings suggest peripheral MCH-levels not to be altered in depression but possibly reflecting depression-related state properties that can be modulated by sleep, medication and sex.
Collapse
Affiliation(s)
- Frank M Schmidt
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany.
| | - Claudia Nowak
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| | - Juergen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany
| | - Christian Sander
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| | - Ulrich Hegerl
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| | - Peter Schönknecht
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| |
Collapse
|
30
|
Transient expression of neuropeptide W in postnatal mouse hypothalamus--a putative regulator of energy homeostasis. Neuroscience 2015; 301:323-37. [PMID: 26073698 DOI: 10.1016/j.neuroscience.2015.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/29/2015] [Accepted: 06/06/2015] [Indexed: 11/24/2022]
Abstract
Neuropeptide B and W (NPB and NPW) are cognate peptide ligands for NPBWR1 (GPR7), a G protein-coupled receptor. In rodents, they have been implicated in the regulation of energy homeostasis, neuroendocrine/autonomic responses, and social interactions. Although localization of these peptides and their receptors in adult rodent brain has been well documented, their expression in mouse brain during development is unknown. Here we demonstrate the transient expression of NPW mRNA in the dorsomedial hypothalamus (DMH) of postnatal mouse brain and its co-localization with neuropeptide Y (NPY) mRNA. Neurons expressing both NPW and NPY mRNAs begin to emerge in the DMH at about postnatal day 0 (P-0) through P-3. Their expression is highest around P-14, declines after P-21, and by P-28 only a faint expression of NPW and NPY mRNA remains. In P-18 brains, we detected NPW neurons in the region spanning the subincertal nucleus (SubI), the lateral hypothalamic (LH) perifornical (PF) areas, and the DMH, where the highest expression of NPW mRNA was observed. The majority of these postnatal hypothalamic NPW neurons co-express NPY mRNA. A cross of NPW-iCre knock-in mice with a Cre-dependent tdTomato reporter line revealed that more than half of the reporter-positive neurons in the adult DMH, which mature from the transiently NPW-expressing neurons, are sensitive to peripherally administrated leptin. These data suggest that the DMH neurons that transiently co-express NPW and NPY in the peri-weaning period might play a role in regulating energy homeostasis during postnatal development.
Collapse
|
31
|
Roth GJ, Heckel A, Kley JT, Lehmann T, Müller SG, Oost T, Rudolf K, Arndt K, Budzinski R, Lenter M, Lotz RRH, Schindler M, Thomas L, Stenkamp D. Design, synthesis and evaluation of MCH receptor 1 antagonists--Part II: Optimization of pyridazines toward reduced phospholipidosis and hERG inhibition. Bioorg Med Chem Lett 2015; 25:3270-4. [PMID: 26077492 DOI: 10.1016/j.bmcl.2015.05.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 10/23/2022]
Abstract
Despite recent success there remains a high therapeutic need for the development of drugs targeting diseases associated with the metabolic syndrome. As part of our search for safe and effective MCH-R1 antagonists for the treatment of obesity, a series of 3,6-disubstituted pyridazines was evaluated. During optimization several issues of the initial lead structures had to be resolved, such as selectivity over related GPCRs, inhibition of the hERG channel as well as the potential to induce phospholipidosis. Utilizing property-based design, we could demonstrate that all parameters can significantly be improved by consequently increasing the polarity of the compounds. By this strategy, we succeeded in identifying potent and orally available MCH-R1 antagonists with good selectivity over M1 and 5-HT2A and an improved safety profile with respect to hERG inhibition and phospholipidosis.
Collapse
Affiliation(s)
- Gerald J Roth
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany.
| | - Armin Heckel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Jörg T Kley
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Thorsten Lehmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Stephan G Müller
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Thorsten Oost
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Klaus Rudolf
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Kirsten Arndt
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Cardiometabolic Research, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Ralph Budzinski
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Cardiometabolic Research, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Martin Lenter
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Cardiometabolic Research, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Ralf R H Lotz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Drug Discovery Support, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Marcus Schindler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Cardiometabolic Research, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Leo Thomas
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Cardiometabolic Research, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Dirk Stenkamp
- Boehringer Ingelheim Pharma GmbH & Co. KG, Department of Medicinal Chemistry, Research Germany, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| |
Collapse
|
32
|
|
33
|
Müller-Fielitz H, Lau M, Geißler C, Werner L, Winkler M, Raasch W. Preventing leptin resistance by blocking angiotensin II AT1 receptors in diet-induced obese rats. Br J Pharmacol 2014; 172:857-68. [PMID: 25258168 DOI: 10.1111/bph.12949] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/03/2014] [Accepted: 09/18/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE AT1 receptor blockers (ARBs) represent an approach for treating metabolic syndrome due to their potency in reducing hypertension, body weight and onset of type 2 diabetes. The mechanism underlying ARB-induced weight loss is still unclear. EXPERIMENTAL APPROACH Leptin resistance tests (LRTs) in diet-induced obese or lean rats were conducted to determine whether telmisartan (8 mg·kg(-1) ·day(-1) , 14 days) enhances leptin sensitivity. Phosphorylation of signal transducer and activator of transcription 3 (pSTAT3) staining was performed in hypothalami to determine leptin transport across the blood-brain barrier. KEY RESULTS Telmisartin reduced weight gain, food intake and plasma leptin but blood pressure remained unchanged. The 24 h profiles of plasma leptin after saline injections were similar in controls and telmisartan-treated rats, but after leptin injections were higher in controls and slightly lower in telmisartan-treated animals. After telmisartan, energy intake during LRT was lower in leptin- than in saline-pretreated rats, but remained unchanged in controls, irrespectively of whether rats received saline or leptin. Leptin minimized the gain in body weight during LRT in telmisartan-treated rats as compared with saline-treated animals. pSTAT3 staining was reduced in cafeteria diet-fed rats as compared with chow-fed rats but this was normalized by telmisartan. Telmisartin reduced hypothalamic mRNA levels of the orexigenic peptides melanin-concentrating hormone and prepro-orexin. CONCLUSIONS AND IMPLICATIONS Rats fed a cafeteria diet develop leptin resistance after 2 weeks. Leptin sensitivity was preserved by telmisartan treatment even in rats fed a cafeteria diet. This pleiotropic effect is not related to the hypotensive action of telmisartan.
Collapse
Affiliation(s)
- Helge Müller-Fielitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | | | | | | | | | | |
Collapse
|
34
|
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
|
35
|
Parks GS, Olivas ND, Ikrar T, Sanathara NM, Wang L, Wang Z, Civelli O, Xu X. Histamine inhibits the melanin-concentrating hormone system: implications for sleep and arousal. J Physiol 2014; 592:2183-96. [PMID: 24639485 DOI: 10.1113/jphysiol.2013.268771] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Melanin-concentrating hormone (MCH)-producing neurons are known to regulate a wide variety of physiological functions such as feeding, metabolism, anxiety and depression, and reward. Recent studies have revealed that MCH neurons receive projections from several wake-promoting brain regions and are integral to the regulation of rapid eye movement (REM) sleep. Here, we provide evidence in both rats and mice that MCH neurons express histamine-3 receptors (H3R), but not histamine-1 (H1R) or histamine-2 (H2R) receptors. Electrophysiological recordings in brain slices from a novel line of transgenic mice that specifically express the reporter ZsGreen in MCH neurons show that histamine strongly inhibits MCH neurons, an effect which is TTX insensitive, and blocked by the intracellular presence of GDP-β-S. A specific H3R agonist, α-methylhistamine, mimicks the inhibitory effects of histamine, and a specific neutral H3R antagonist, VUF 5681, blocks this effect. Tertiapin Q (TPQ), a G protein-dependent inwardly rectifying potassium (GIRK) channel inhibitor, abolishes histaminergic inhibition of MCH neurons. These results indicate that histamine directly inhibits MCH neurons through H3R by activating GIRK channels and suggest that that inhibition of the MCH system by wake-active histaminergic neurons may be responsible for silencing MCH neurons during wakefulness and thus may be directly involved in the regulation of sleep and arousal.
Collapse
Affiliation(s)
- Gregory S Parks
- Department of Pharmacology, University of California Irvine, Irvine, CA, 92697, USA Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697, USA
| | - Nicholas D Olivas
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, 92697, USA
| | - Taruna Ikrar
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, 92697, USA
| | - Nayna M Sanathara
- Department of Pharmacology, University of California Irvine, Irvine, CA, 92697, USA
| | - Lien Wang
- Department of Pharmacology, University of California Irvine, Irvine, CA, 92697, USA
| | - Zhiwei Wang
- Department of Pharmacology, University of California Irvine, Irvine, CA, 92697, USA
| | - Olivier Civelli
- Department of Pharmacology, University of California Irvine, Irvine, CA, 92697, USA Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697, USA Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, 92697, USA
| |
Collapse
|
36
|
Chee MJS, Pissios P, Prasad D, Maratos-Flier E. Expression of melanin-concentrating hormone receptor 2 protects against diet-induced obesity in male mice. Endocrinology 2014; 155:81-8. [PMID: 24169555 PMCID: PMC3868808 DOI: 10.1210/en.2013-1738] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
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.
Collapse
Affiliation(s)
- Melissa J S Chee
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | | | | | | |
Collapse
|
37
|
Motani AS, Luo J, Liang L, Mihalic JT, Chen X, Tang L, Li L, Jaen J, Chen JL, Dai K. Evaluation of AMG 076, a potent and selective MCHR1 antagonist, in rodent and primate obesity models. Pharmacol Res Perspect 2013; 1:e00003. [PMID: 25505557 PMCID: PMC4184568 DOI: 10.1002/prp2.3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 07/02/2013] [Accepted: 07/04/2013] [Indexed: 02/06/2023] Open
Abstract
Melanin-concentrating hormone (MCH) regulates food intake through activation of the receptor, MCHR1. We have identified AMG 076 as an orally bioavailable potent and selective small molecule antagonist of MCHR1. In mouse models of obesity, AMG 076 caused a reduction in body weight gain in wild-type (MCHR1+/+) but not in knockout (MCHR1−/−) mice. The body weight reduction was associated with decreases in food intake and increases in energy expenditure. Importantly, we show that these MCHR1-dependent effects of AMG 076 were also reflected in improved metabolic phenotypes, increased glucose tolerance and insulin sensitivity. Preliminary data on effects of AMG 076 in obese cynomolgus monkeys are also presented.
Collapse
|
38
|
Kátai Z, Adori C, Kitka T, Vas S, Kalmár L, Kostyalik D, Tóthfalusi L, Palkovits M, Bagdy G. Acute escitalopram treatment inhibits REM sleep rebound and activation of MCH-expressing neurons in the lateral hypothalamus after long term selective REM sleep deprivation. Psychopharmacology (Berl) 2013; 228:439-49. [PMID: 23515582 DOI: 10.1007/s00213-013-3046-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 02/25/2013] [Indexed: 01/28/2023]
Abstract
RATIONALE Selective rapid eye movement sleep (REMS) deprivation using the platform-on-water ("flower pot") method causes sleep rebound with increased REMS, decreased REMS latency, and activation of the melanin-concentrating hormone (MCH) expressing neurons in the hypothalamus. MCH is implicated in the pathomechanism of depression regarding its influence on mood, feeding behavior, and REMS. OBJECTIVES We investigated the effects of the most selective serotonin reuptake inhibitor escitalopram on sleep rebound following REMS deprivation and, in parallel, on the activation of MCH-containing neurons. METHODS Escitalopram or vehicle (10 mg/kg, intraperitoneally) was administered to REMS-deprived (72 h) or home cage male Wistar rats. During the 3-h-long "rebound sleep", electroencephalography was recorded, followed by an MCH/Fos double immunohistochemistry. RESULTS During REMS rebound, the time spent in REMS and the number of MCH/Fos double-labeled neurons in the lateral hypothalamus increased markedly, and REMS latency showed a significant decrease. All these effects of REMS deprivation were significantly attenuated by escitalopram treatment. Besides the REMS-suppressing effects, escitalopram caused an increase in amount of and decrease in latency of slow wave sleep during the rebound. CONCLUSIONS These results show that despite the high REMS pressure caused by REMS deprivation procedure, escitalopram has the ability to suppress REMS rebound, as well as to diminish the activation of MCH-containing neurons, in parallel. Escitalopram caused a shift from REMS to slow wave sleep during the rebound. Furthermore, these data point to the potential connection between the serotonergic system and MCH in sleep regulation, which can be relevant in depression and in other mood disorders.
Collapse
Affiliation(s)
- Zita Kátai
- Department of Pharmacodynamics, Semmelweis University, 1089 Nagyvárad tér 4., Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Nagata A, Hamamoto A, Horikawa M, Yoshimura K, Takeda S, Saito Y. Characterization of ciliary targeting sequence of rat melanin-concentrating hormone receptor 1. Gen Comp Endocrinol 2013; 188:159-65. [PMID: 23467069 DOI: 10.1016/j.ygcen.2013.02.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 02/14/2013] [Accepted: 02/19/2013] [Indexed: 10/27/2022]
Abstract
Melanin-concentrating hormone (MCH) is the natural peptide ligand for MCHR1 and MCHR2, which belong to the G protein-coupled receptor (GPCR) superfamily. The MCH-MCHR1 system is involved in the regulation of feeding, energy homeostasis and emotional processing in rodents. Recently, MCHR1 expression was discovered in neuronal immotile primary cilia of the central nervous system in mice. The cilium has an important chemosensory function in many types of cell and ciliary dysfunction is associated with cliopathies such as polycystic kidney disease, retinal dystrophy, and obesity. The targeting sequence of ciliary membrane proteins is thought to be unique. Although these sequences have been predicted in the cytoplasmic third loop and/or C-terminus of GPCRs, little is known about the characteristics of MCHR1. We thus explored the molecular mechanisms of MCHR1 targeting by transiently expressing a series of MCHR1 mutants into ciliated hRPE1 cells and evaluated the effects of these mutations on the ciliary localization of the heterologous receptor. This approach demonstrated that an Ala-to-Gly mutation (A242G) within the third intracellular loop induced a significant reduction in ciliary localization of the receptor without affecting the ciliogenesis. In contrast, no C-terminal truncation mutant had any effect on ciliary localization or cilia length. This study provides a potential molecular link between defective cilia and clinical manifestations such as obesity.
Collapse
Affiliation(s)
- Asami Nagata
- Graduate School of Integrated Arts and Sciences, Hiroshima University, Kagamiyama, Hiroshima 739-8521, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Ziogas DC, Gras-Miralles B, Mustafa S, Geiger BM, Najarian RM, Nagel JM, Flier SN, Popov Y, Tseng YH, Kokkotou E. Anti-melanin-concentrating hormone treatment attenuates chronic experimental colitis and fibrosis. Am J Physiol Gastrointest Liver Physiol 2013; 304:G876-84. [PMID: 23538494 PMCID: PMC3652072 DOI: 10.1152/ajpgi.00305.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibrosis represents a major complication of several chronic diseases, including inflammatory bowel disease (IBD). Treatment of IBD remains a clinical challenge despite several recent therapeutic advances. Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide shown to regulate appetite and energy balance. However, accumulating evidence suggests that MCH has additional biological effects, including modulation of inflammation. In the present study, we examined the efficacy of an MCH-blocking antibody in treating established, dextran sodium sulfate-induced experimental colitis. Histological and molecular analysis of mouse tissues revealed that mice receiving anti-MCH had accelerated mucosal restitution and lower colonic expression of several proinflammatory cytokines, as well as fibrogenic genes, including COL1A1. In parallel, they spared collagen deposits seen in the untreated mice, suggesting attenuated fibrosis. These findings raised the possibility of perhaps direct effects of MCH on myofibroblasts. Indeed, in biopsies from patients with IBD, we demonstrate expression of the MCH receptor MCHR1 in α-smooth muscle actin(+) subepithelial cells. CCD-18Co cells, a primary human colonic myofibroblast cell line, were also positive for MCHR1. In these cells, MCH acted as a profibrotic modulator by potentiating the effects of IGF-1 and TGF-β on proliferation and collagen production. Thus, by virtue of combined anti-inflammatory and anti-fibrotic effects, blocking MCH might represent a compelling approach for treating IBD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yury Popov
- 1Beth Israel Deaconess Medical Center and
| | - Yu-Hua Tseng
- 2Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
41
|
Schmidt FM, Kratzsch J, Gertz HJ, Tittmann M, Jahn I, Pietsch UC, Kaisers UX, Thiery J, Hegerl U, Schönknecht P. Cerebrospinal fluid melanin-concentrating hormone (MCH) and hypocretin-1 (HCRT-1, orexin-A) in Alzheimer's disease. PLoS One 2013; 8:e63136. [PMID: 23667582 PMCID: PMC3646736 DOI: 10.1371/journal.pone.0063136] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 03/28/2013] [Indexed: 11/24/2022] Open
Abstract
Ancillary to decline in cognitive abilities, patients with Alzheimer’s disease (AD) frequently suffer from behavioural and psychological symptoms of dementia (BPSD). Hypothalamic polypeptides such as melanin-concentrating hormone (MCH) and hypocretin-1 (HCRT-1, orexin-A) are promoters of sleep-wake regulation and energy homeostasis and are found to impact on cognitive performance. To investigate the role of MCH and HCRT-1 in AD, cerebrospinal fluid (CSF) levels were measured in 33 patients with AD and 33 healthy subjects (HS) using a fluorescence immunoassay (FIA). A significant main effect of diagnosis (F(1,62) = 8.490, p<0.01) on MCH levels was found between AD (93.76±13.47 pg/mL) and HS (84.65±11.40 pg/mL). MCH correlated with T-tau (r = 0.47; p<0.01) and P-tau (r = 0.404; p<0.05) in the AD but not in the HS. CSF-MCH correlated negatively with MMSE scores in the AD (r = −0.362, p<0.05) and was increased in more severely affected patients (MMSE≤20) compared to HS (p<0.001) and BPSD-positive patients compared to HS (p<0.05). In CSF-HCRT-1, a significant main effect of sex (F(1,31) = 4.400, p<0.05) with elevated levels in females (90.93±17.37 pg/mL vs. 82.73±15.39 pg/mL) was found whereas diagnosis and the sex*diagnosis interaction were not significant. Elevated levels of MCH in patients suffering from AD and correlation with Tau and severity of cognitive impairment point towards an impact of MCH in AD. Gender differences of CSF-HCRT-1 controversially portend a previously reported gender dependence of HCRT-1-regulation. Histochemical and actigraphic explorations are warranted to further elucidate alterations of hypothalamic transmitter regulation in AD.
Collapse
Affiliation(s)
- Frank M Schmidt
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hopf FW, Seif T, Chung S, Civelli O. MCH and apomorphine in combination enhance action potential firing of nucleus accumbens shell neurons in vitro. PeerJ 2013; 1:e61. [PMID: 23646281 PMCID: PMC3642701 DOI: 10.7717/peerj.61] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/12/2013] [Indexed: 11/20/2022] Open
Abstract
The MCH and dopamine receptor systems have been shown to modulate a number of behaviors related to reward processing, addiction, and neuropsychiatric conditions such as schizophrenia and depression. In addition, MCH and dopamine receptors can interact in a positive manner, for example in the expression of cocaine self-administration. A recent report (Chung et al., 2011a) showed that the DA1/DA2 dopamine receptor activator apomorphine suppresses pre-pulse inhibition, a preclinical model for some aspects of schizophrenia. Importantly, MCH can enhance the effects of lower doses of apomorphine, suggesting that co-modulation of dopamine and MCH receptors might alleviate some symptoms of schizophrenia with a lower dose of dopamine receptor modulator and thus fewer potential side effects. Here, we investigated whether MCH and apomorphine could enhance action potential firing in vitro in the nucleus accumbens shell (NAshell), a region which has previously been shown to mediate some behavioral effects of MCH. Using whole-cell patch-clamp electrophysiology, we found that MCH, which has no effect on firing on its own, was able to increase NAshell firing when combined with a subthreshold dose of apomorphine. Further, this MCH/apomorphine increase in firing was prevented by an antagonist of either a DA1 or a DA2 receptor, suggesting that apomorphine acts through both receptor types to enhance NAshell firing. The MCH/apomorphine-mediated firing increase was also prevented by an MCH receptor antagonist or a PKA inhibitor. Taken together, our results suggest that MCH can interact with lower doses of apomorphine to enhance NAshell firing, and thus that MCH and apomorphine might interact in vivo within the NAshell to suppress pre-pulse inhibition.
Collapse
Affiliation(s)
- F Woodward Hopf
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California , San Francisco, Emeryville, CA , USA
| | | | | | | |
Collapse
|
43
|
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
|
44
|
Conductier G, Martin AO, Risold PY, Jego S, Lavoie R, Lafont C, Mollard P, Adamantidis A, Nahon JL. Control of ventricular ciliary beating by the melanin concentrating hormone-expressing neurons of the lateral hypothalamus: a functional imaging survey. Front Endocrinol (Lausanne) 2013; 4:182. [PMID: 24324458 PMCID: PMC3839296 DOI: 10.3389/fendo.2013.00182] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/07/2013] [Indexed: 12/26/2022] Open
Abstract
The cyclic peptide Melanin Concentrating Hormone (MCH) is known to control a large number of brain functions in mammals such as food intake and metabolism, stress response, anxiety, sleep/wake cycle, memory, and reward. Based on neuro-anatomical and electrophysiological studies these functions were attributed to neuronal circuits expressing MCHR1, the single MCH receptor in rodents. In complement to our recently published work (1) we provided here new data regarding the action of MCH on ependymocytes in the mouse brain. First, we establish that MCHR1 mRNA is expressed in the ependymal cells of the third ventricle epithelium. Second, we demonstrated a tonic control of MCH-expressing neurons on ependymal cilia beat frequency using in vitro optogenics. Finally, we performed in vivo measurements of CSF flow using fluorescent micro-beads in wild-type and MCHR1-knockout mice. Collectively, our results demonstrated that MCH-expressing neurons modulate ciliary beating of ependymal cells at the third ventricle and could contribute to maintain cerebro-spinal fluid homeostasis.
Collapse
Affiliation(s)
- Grégory Conductier
- UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Valbonne, France
- University of Nice Sophia Antipolis, Nice, France
| | - Agnès O. Martin
- UMR5203, Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Montpellier, France
- U661, INSERM, Montpellier, France
- UMR-5203, Universités de Montpellier 1 & 2, Montpellier, France
| | - Pierre-Yves Risold
- Laboratoire d’Histologie, IFR 133, Faculté de Médecine et de Pharmacie, Besançon, France
| | - Sonia Jego
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Raphaël Lavoie
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Chrystel Lafont
- UMR5203, Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Montpellier, France
- U661, INSERM, Montpellier, France
- UMR-5203, Universités de Montpellier 1 & 2, Montpellier, France
| | - Patrice Mollard
- UMR5203, Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Montpellier, France
- U661, INSERM, Montpellier, France
- UMR-5203, Universités de Montpellier 1 & 2, Montpellier, France
| | | | - Jean-Louis Nahon
- UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Valbonne, France
- University of Nice Sophia Antipolis, Nice, France
- Station de Primatologie, UPS 846, Centre National de la Recherche Scientifique, Rousset sur Arc, France
- *Correspondence: Jean-Louis Nahon, UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, 660 Route des Lucioles, Sophia Antipolis, Valbonne, France e-mail:
| |
Collapse
|
45
|
Distribution of secretin receptors in the rat central nervous system: an in situ hybridization study. J Mol Neurosci 2012; 50:172-8. [PMID: 23065333 DOI: 10.1007/s12031-012-9895-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022]
Abstract
Secretin shows a wide distribution in the brain. Functional significance of central secretin is stressed since it has been associated with autism and schizophrenia. The presence of the secretin receptor was previously demonstrated in the brain by different methods. Neurons in the cerebellum, hypothalamic paraventricular and supraoptic nuclei, and in the vascular organ of lamina terminalis were shown to express secretin receptor mRNA by using in situ hybridization with digoxigenin-labeled probe. In this work, we used a very sensitive radioactive in situ hybridization technique and systematically mapped the expression of secretin receptor mRNA in the brain. The densest labeling was observed in the nucleus of solitary tract and in the laterodorsal thalamic nucleus, where decreasing number of receptors was seen in the vascular organ of lamina terminalis, and the lateral habenular complex, and then in the supraoptic nucleus. Only a few scattered labeled cells were observed in the median frontal gyrus, entorhinal cortex, hypothalamic paraventricular nucleus, perifornical region, lateral hypothalamic area, head of the caudate nucleus, spinal trigeminal nucleus, and cerebellum. Secretin receptor mRNA showed a far wider distribution than was known before, suggesting a more significant functional relevance than thought earlier.
Collapse
|
46
|
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]
|
47
|
Heteromerization of ciliary G protein-coupled receptors in the mouse brain. PLoS One 2012; 7:e46304. [PMID: 23029470 PMCID: PMC3459911 DOI: 10.1371/journal.pone.0046304] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/30/2012] [Indexed: 11/22/2022] Open
Abstract
Nearly every cell type in the mammalian body projects from its cell surface a primary cilium that provides important sensory and signaling functions. Defects in the formation or function of primary cilia have been implicated in the pathogenesis of many human developmental disorders and diseases, collectively termed ciliopathies. Most neurons in the brain possess cilia that are enriched for signaling proteins such as G protein-coupled receptors and adenylyl cyclase type 3, suggesting neuronal cilia sense neuromodulators in the brain and contribute to non-synaptic signaling. Indeed, disruption of neuronal cilia or loss of neuronal ciliary signaling proteins is associated with obesity and learning and memory deficits. As the functions of primary cilia are defined by the signaling proteins that localize to the ciliary compartment, identifying the complement of signaling proteins in cilia can provide important insights into their physiological roles. Here we report for the first time that different GPCRs can colocalize within the same cilium. Specifically, we found the ciliary GPCRs, melanin-concentrating hormone receptor 1 (Mchr1) and somatostatin receptor 3 (Sstr3) colocalizing within cilia in multiple mouse brain regions. In addition, we have evidence suggesting Mchr1 and Sstr3 form heteromers. As GPCR heteromerization can affect ligand binding properties as well as downstream signaling, our findings add an additional layer of complexity to neuronal ciliary signaling.
Collapse
|
48
|
García-Fuster MJ, Parks GS, Clinton SM, Watson SJ, Akil H, Civelli O. The melanin-concentrating hormone (MCH) system in an animal model of depression-like behavior. Eur Neuropsychopharmacol 2012; 22:607-13. [PMID: 22209364 PMCID: PMC3319808 DOI: 10.1016/j.euroneuro.2011.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/20/2011] [Accepted: 12/02/2011] [Indexed: 02/07/2023]
Abstract
Selective breeding for divergence in locomotion in a novel environment (bHR, bred High-Responder; bLR, bred Low-Responder) correlates with stress-reactivity, spontaneous anxiety-like behaviors and predicts vulnerability in a rodent model of depression. Identifying genetic factors that may account for such vulnerability are key determinants not only for the illness outcome but also for the development of better-tailored treatment options. Melanin-concentrating hormone (MCH) is a neuropeptide that exhibits some of the hallmarks of a regulator of affective states. The aim of this study was to ascertain the role of the MCH system in depression-like behaviors in bHR vs. bLR rats. bLR rats showed a 44% increase in hypothalamic pMCH mRNA and a 14% decrease in hippocampal CA1 MCH1R mRNA when compared to bHR rats. Interestingly, the amount of time that rats spent immobile in the FST (depressive-like behavior) correlated positively with the amount of hypothalamic pMCH mRNA and negatively with that of hippocampal CA1 MCH1R. The results indicate that the bLR-bHR is a useful rat model to investigate individual basal genetic differences that participate in the monitoring of emotional responsiveness (i.e., depression- and anxiety-like behaviors). They also point to the MCH system (i.e., chronically higher pMCH expression and consequently receptor down-regulation) as a candidate biomarker for the severity of depressive-like behavior. The data indicate that MCH1R participates in the modulation of depression-like behavior through a process that involves the CA1 region of the hippocampus, supporting the possible use of MCH1R antagonists in the treatment of depression.
Collapse
Affiliation(s)
- M J García-Fuster
- Department of Pharmacology, University of California, Irvine, CA, United States.
| | | | | | | | | | | |
Collapse
|
49
|
Nagel JM, Geiger BM, Karagiannis AKA, Gras-Miralles B, Horst D, Najarian RM, Ziogas DC, Chen X, Kokkotou E. Reduced intestinal tumorigenesis in APCmin mice lacking melanin-concentrating hormone. PLoS One 2012; 7:e41914. [PMID: 22848656 PMCID: PMC3407051 DOI: 10.1371/journal.pone.0041914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/27/2012] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Melanin-concentrating hormone (MCH) is an evolutionary conserved hypothalamic neuropeptide that in mammals primarily regulates appetite and energy balance. We have recently identified a novel role for MCH in intestinal inflammation by demonstrating attenuated experimental colitis in MCH deficient mice or wild type mice treated with an anti-MCH antibody. Therefore, targeting MCH has been proposed for the treatment of inflammatory bowel disease. Given the link between chronic intestinal inflammation and colorectal cancer, in the present study we sought to investigate whether blocking MCH might have effects on intestinal tumorigenesis that are independent of inflammation. METHODOLOGY Tumor development was evaluated in MCH-deficient mice crossed to the APCmin mice which develop spontaneously intestinal adenomas. A different cohort of MCH-/- and MCH+/+ mice in the APCmin background was treated with dextran sodium sulphate (DSS) to induce inflammation-dependent colorectal tumors. In Caco2 human colorectal adenocarcinoma cells, the role of MCH on cell survival, proliferation and apoptosis was investigated. RESULTS APCmin mice lacking MCH developed fewer, smaller and less dysplastic tumors in the intestine and colon which at the molecular level are characterized by attenuated activation of the wnt/beta-catenin signaling pathway and increased apoptotic indices. Form a mechanistic point of view, MCH increased the survival of colonic adenocarcinoma Caco2 cells via inhibiting apoptosis, consistent with the mouse studies. CONCLUSION In addition to modulating inflammation, MCH was found to promote intestinal tumorigenesis at least in part by inhibiting epithelial cell apoptosis. Thereby, blocking MCH as a therapeutic approach is expected to decrease the risk for colorectal cancer.
Collapse
Affiliation(s)
- Jutta M. Nagel
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brenda M. Geiger
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Apostolos K. A. Karagiannis
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Beatriz Gras-Miralles
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David Horst
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert M. Najarian
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dimitrios C. Ziogas
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - XinHua Chen
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Efi Kokkotou
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
50
|
Melanin-concentrating hormone receptor 1 (MCH1-R) antagonism: reduced appetite for calories and suppression of addictive-like behaviors. Pharmacol Biochem Behav 2012; 102:400-6. [PMID: 22705492 DOI: 10.1016/j.pbb.2012.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 05/29/2012] [Accepted: 06/09/2012] [Indexed: 11/23/2022]
Abstract
RATIONALE The hypothalamic neuropeptide melanin-concentrating hormone and its MCH1 receptor have been implicated in regulation of feeding and energy homeostasis, as well as modulation of reward-related behaviors. Here, we examined whether the MCH system plays a role both in caloric and motivational aspects of sugar intake. MATERIALS AND METHODS The non-peptide MCH1-R antagonist GW803430 (3, 10, 30 mg/kg, i.p.) was first tested on self-administration under a fixed ratio schedule of reinforcement of both a caloric (10% w/v sucrose) and a non-caloric (0.06% w/v saccharin) sweet solution. GW803430 was then tested for its ability to alter motivational properties and seeking of sucrose. Lastly, the drug was tested to concurrently examine its effects on the escalated consumption of both sugar and food in animals following intermittent sugar access. RESULTS The MCH1-R antagonist reduced sucrose- but not saccharin-reinforced lever pressing, likely reflecting a decreased appetite for calories in GW803430-treated rats. GW803430 reduced sucrose self-administration under a progressive ratio schedule, and suppressed cue-induced reinstatement of sucrose seeking, suggesting effects on rewarding properties of sucrose. GW803430 attenuated food intake in rats on intermittent access to sucrose at all doses examined (3, 10, 30 mg/kg), while reduction of sugar intake was weaker in magnitude. CONCLUSION Together, these observations support an involvement of the MCH system in regulation of energy balance as well as mediation of sucrose reward. MCH may be an important regulator of sugar intake by acting on both caloric and rewarding components.
Collapse
|