1
|
Shi L, He Y, Lian Y, Luo J, Zhu X, Zhao H. Melanin-concentrating hormone: A promising target for antidepressant treatment. Pharmacol Biochem Behav 2025; 250:173999. [PMID: 40081601 DOI: 10.1016/j.pbb.2025.173999] [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: 12/08/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Depression represents a complex neuropsychiatric disorder with an escalating global health burden, characterized by heterogeneous pathophysiology and profound impairments in cognitive-emotional functioning. Current treatment methods have limited efficacy in some individuals and may induce undesirable side effects, necessitating the exploration of novel therapeutic targets and techniques. Emerging research has identified neuropeptide systems as pivotal regulators of mood-related circuits, with melanin-concentrating hormone (MCH) signaling emerging as a particularly promising candidate for antidepressant development. The potential involvement of MCH in the pathophysiology of depression was first proposed over two decades ago. Since then, accumulating evidence from recent studies has progressively illuminated its multifaceted roles in modulating depressive behaviors and underlying neurobiological mechanisms. This review systematically analyzes the mechanistic interplay between MCH signaling and depression pathophenotypes, including its relationship with the hypothalamic-pituitary-adrenal (HPA) axis, neurotransmitter systems, synaptic plasticity, and the regulation of sleep-wakefulness. Particular emphasis is placed on advancing the therapeutic rationale for MCH receptor 1 (MCHR1) antagonists, which demonstrate rapid-onset antidepressant efficacy in preclinical studies compared to traditional agents. Nonetheless, the antidepressant mechanism of the MCH system still requires further elucidation to confirm its therapeutic potential.
Collapse
Affiliation(s)
- Lingchang Shi
- School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Ying He
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan 410208, China
| | - Yujun Lian
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jie Luo
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xuan Zhu
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan 410208, China; Department of Science & Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan 410208, China.
| |
Collapse
|
2
|
Chaki S. Melanin-concentrating hormone receptor: A therapeutic target for novel anxiolytics. Pharmacol Biochem Behav 2024; 242:173818. [PMID: 38971471 DOI: 10.1016/j.pbb.2024.173818] [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: 05/30/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Anxiety disorders are chronic, disabling psychiatric disorders, and there is a growing medical need for the development of novel pharmacotherapeutic agents showing improved efficacy and an improved side effect profile as compared with the currently prescribed anxiolytic drugs. In the course of the search for next-generation anxiolytics, neuropeptide receptors have garnered interest as potential therapeutic targets, underscored by pivotal roles in modulating stress responses and findings from animal studies using pharmacological tools. Among these neuropeptide receptors, the type 1 receptor for melanin-concentrating hormone (MCH1), which has been demonstrated to be involved in an array of physiological processes, including the regulation of stress responses and affective states, has gained attraction as a therapeutic target for drugs used in the treatment of psychiatric disorders, including anxiety disorders. To date, a plethora of MCH1 antagonists have been synthesized, and studies using MCH1 antagonists and genetically manipulated mice lacking MCH1 have revealed that the blockade of MCH1 produces anxiolytic-like effects across diverse rodent paradigms. In addition, MCH1 antagonists have been demonstrated to show a rapid onset of antidepressant-like effects; therefore, they may be effective for conditions commonly encountered in patients with anxiety disorders, which is an advantage for anxiolytic drugs. Notably, MCH1 antagonists have not manifested the undesirable side effects observed with the currently prescribed anxiolytics. All these preclinical findings testify to the potential of MCH1 antagonists as novel anxiolytics. Although there are still issues that need to be resolved prior to the initiation of clinical trials, such as elucidating the precise neuronal mechanisms underlying their anxiolytic effects and exploring pertinent biomarkers that can be used in clinical trials, MCH1 blockade appears to be an attractive way to tackle anxiety disorders.
Collapse
Affiliation(s)
- Shigeyuki Chaki
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Saitama 331-9530, Japan; Chiba University Center for Forensic Mental Health, Chiba, Chiba 260-8670, Japan.
| |
Collapse
|
3
|
Abstract
OBJECTIVE Due to the phenotypic heterogeneity and etiological complexity of bipolar disorder (BD), many patients do not respond well to the current medications, and developing novel effective treatment is necessary. Whether any BD genome-wide association study (GWAS) risk genes were targets of existing drugs or novel drugs that can be repurposed in the clinical treatment of BD is a hot topic in the GWAS era of BD. METHODS A list of 425 protein-coding BD risk genes was distilled through the BD GWAS, and 4479 protein-coding druggable targets were retrieved from the druggable genome. The overlapped genes/targets were subjected to further analyses in DrugBank, Pharos, and DGIdb datasets in terms of their FDA status, mechanism of action and primary indication, to identify their potential for repurposing. RESULTS We identified 58 BD GWAS risk genes grouped as the druggable targets, and several genes were given higher priority. These BD risk genes were targets of antipsychotics, antidepressants, antiepileptics, calcium channel antagonists, as well as anxiolytics and analgesics, either existing clinically-approved drugs for BD or the drugs than can be repurposed for treatment of BD in the future. Those genes were also likely relevant to BD pathophysiology, as many of them encode ion channel, ion transporter or neurotransmitter receptor, or the mice manipulating those genes are likely to mimic the phenotypes manifest in BD patients. CONCLUSIONS This study identifies several targets that may facilitate the discovery of novel treatments in BD, and implies the value of conducting GWAS into clinical translation.
Collapse
Affiliation(s)
- Hao-Xiang Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
4
|
Gajjala RR, Chinta RR, Gopireddy VSR, Poola S, Balam SK, Chintha V, Pasupuleti VR, Avula VKR, Vallela S, Vasilievich Zyryanov G, Cirandur SR. Ethyl-4-(aryl)-6-methyl-2-(oxo/thio)-3,4-dihydro-1H-pyrimidine-5-carboxylates: Silica supported bismuth(III)triflate catalyzed synthesis and antioxidant activity. Bioorg Chem 2022; 129:106205. [DOI: 10.1016/j.bioorg.2022.106205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/02/2022]
|
5
|
Potter LE, Burgess CR. The melanin-concentrating hormone system as a target for the treatment of sleep disorders. Front Neurosci 2022; 16:952275. [PMID: 36177357 PMCID: PMC9513178 DOI: 10.3389/fnins.2022.952275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Given the widespread prevalence of sleep disorders and their impacts on health, it is critical that researchers continue to identify and evaluate novel avenues of treatment. Recently the melanin-concentrating hormone (MCH) system has attracted commercial and scientific interest as a potential target of pharmacotherapy for sleep disorders. This interest emerges from basic scientific research demonstrating a role for MCH in regulating sleep, and particularly REM sleep. In addition to this role in sleep regulation, the MCH system and the MCH receptor 1 (MCHR1) have been implicated in a wide variety of other physiological functions and behaviors, including feeding/metabolism, reward, anxiety, depression, and learning. The basic research literature on sleep and the MCH system, and the history of MCH drug development, provide cause for both skepticism and cautious optimism about the prospects of MCH-targeting drugs in sleep disorders. Extensive efforts have focused on developing MCHR1 antagonists for use in obesity, however, few of these drugs have advanced to clinical trials, and none have gained regulatory approval. Additional basic research will be needed to fully characterize the MCH system’s role in sleep regulation, for example, to fully differentiate between MCH-neuron and peptide/receptor-mediated functions. Additionally, a number of issues relating to drug design will continue to pose a practical challenge for novel pharmacotherapies targeting the MCH system.
Collapse
Affiliation(s)
- Liam E. Potter
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Liam E. Potter,
| | - Christian R. Burgess
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- Christian R. Burgess,
| |
Collapse
|
6
|
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
|
7
|
Phenotypes, mechanisms and therapeutics: insights from bipolar disorder GWAS findings. Mol Psychiatry 2022; 27:2927-2939. [PMID: 35351989 DOI: 10.1038/s41380-022-01523-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 12/25/2022]
Abstract
Genome-wide association studies (GWAS) have reported substantial genomic loci significantly associated with clinical risk of bipolar disorder (BD), and studies combining techniques of genetics, neuroscience, neuroimaging, and pharmacology are believed to help tackle clinical problems (e.g., identifying novel therapeutic targets). However, translating findings of psychiatric genetics into biological mechanisms underlying BD pathogenesis remains less successful. Biological impacts of majority of BD GWAS risk loci are obscure, and the involvement of many GWAS risk genes in this illness is yet to be investigated. It is thus necessary to review the progress of applying BD GWAS risk genes in the research and intervention of the disorder. A comprehensive literature search found that a number of such risk genes had been investigated in cellular or animal models, even before they were highlighted in BD GWAS. Intriguingly, manipulation of many BD risk genes (e.g., ANK3, CACNA1C, CACNA1B, HOMER1, KCNB1, MCHR1, NCAN, SHANK2 etc.) resulted in altered murine behaviors largely restoring BD clinical manifestations, including mania-like symptoms such as hyperactivity, anxiolytic-like behavior, as well as antidepressant-like behavior, and these abnormalities could be attenuated by mood stabilizers. In addition to recapitulating phenotypic characteristics of BD, some GWAS risk genes further provided clues for the neurobiology of this illness, such as aberrant activation and functional connectivity of brain areas in the limbic system, and modulated dendritic spine morphogenesis as well as synaptic plasticity and transmission. Therefore, BD GWAS risk genes are undoubtedly pivotal resources for modeling this illness, and might be translational therapeutic targets in the future clinical management of BD. We discuss both promising prospects and cautions in utilizing the bulk of useful resources generated by GWAS studies. Systematic integrations of findings from genetic and neuroscience studies are called for to promote our understanding and intervention of BD.
Collapse
|
8
|
Exploring the role of neuropeptides in depression and anxiety. Prog Neuropsychopharmacol Biol Psychiatry 2022; 114:110478. [PMID: 34801611 DOI: 10.1016/j.pnpbp.2021.110478] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 12/24/2022]
Abstract
Depression is one of the most prevalent forms of mental disorders and is the most common cause of disability in the Western world. Besides, the harmful effects of stress-related mood disorders on the patients themselves, they challenge the health care system with enormous social and economic impacts. Due to the high proportion of patients not responding to existing drugs, finding new treatment strategies has become an important topic in neurobiology, and there is much evidence that neuropeptides are not only involved in the physiology of stress but may also be clinically important. Based on preclinical trial data, new neuropharmaceutical candidates may target neuropeptides and their receptors and are expected to be essential and valuable tools in the treatment of psychiatric disorders. In the current article, we have summarized data obtained from animal models of depressive disorder and transgenic mouse models. We also focus on previously published research data of clinical studies on corticotropin-releasing hormone (CRH), galanin (GAL), neuropeptide Y (NPY), neuropeptide S (NPS), Oxytocin (OXT), vasopressin (VP), cholecystokinin (CCK), and melanin-concentrating hormone (MCH) stress research fields.
Collapse
|
9
|
Myslivecek J. Social Isolation: How Can the Effects on the Cholinergic System Be Isolated? Front Pharmacol 2021; 12:716460. [PMID: 34916930 PMCID: PMC8670609 DOI: 10.3389/fphar.2021.716460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/15/2021] [Indexed: 01/31/2023] Open
Abstract
Social species form organizations that support individuals because the consequent social behaviors help these organisms survive. The isolation of these individuals may be a stressor. We reviewed the potential mechanisms of the effects of social isolation on cholinergic signaling and vice versa how changes in cholinergic signaling affect changes due to social isolation.There are two important problems regarding this topic. First, isolation schemes differ in their duration (1–165 days) and initiation (immediately after birth to adulthood). Second, there is an important problem that is generally not considered when studying the role of the cholinergic system in neurobehavioral correlates: muscarinic and nicotinic receptor subtypes do not differ sufficiently in their affinity for orthosteric site agonists and antagonists. Some potential cholinesterase inhibitors also affect other targets, such as receptors or other neurotransmitter systems. Therefore, the role of the cholinergic system in social isolation should be carefully considered, and multiple receptor systems may be involved in the central nervous system response, although some subtypes are involved in specific functions. To determine the role of a specific receptor subtype, the presence of a specific subtype in the central nervous system should be determined using search in knockout studies with the careful application of specific agonists/antagonists.
Collapse
Affiliation(s)
- Jaromir Myslivecek
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
10
|
Bicks LK, Koike H, Akbarian S, Morishita H. Prefrontal Cortex and Social Cognition in Mouse and Man. Front Psychol 2015; 6:1805. [PMID: 26635701 PMCID: PMC4659895 DOI: 10.3389/fpsyg.2015.01805] [Citation(s) in RCA: 346] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/09/2015] [Indexed: 12/15/2022] Open
Abstract
Social cognition is a complex process that requires the integration of a wide variety of behaviors, including salience, reward-seeking, motivation, knowledge of self and others, and flexibly adjusting behavior in social groups. Not surprisingly, social cognition represents a sensitive domain commonly disrupted in the pathology of a variety of psychiatric disorders including Autism Spectrum Disorder (ASD) and Schizophrenia (SCZ). Here, we discuss convergent research from animal models to human disease that implicates the prefrontal cortex (PFC) as a key regulator in social cognition, suggesting that disruptions in prefrontal microcircuitry play an essential role in the pathophysiology of psychiatric disorders with shared social deficits. We take a translational perspective of social cognition, and review three key behaviors that are essential to normal social processing in rodents and humans, including social motivation, social recognition, and dominance hierarchy. A shared prefrontal circuitry may underlie these behaviors. Social cognition deficits in animal models of neurodevelopmental disorders like ASD and SCZ have been linked to an altered balance of excitation and inhibition (E/I ratio) within the cortex generally, and PFC specifically. A clear picture of the mechanisms by which altered E/I ratio in the PFC might lead to disruptions of social cognition across a variety of behaviors is not well understood. Future studies should explore how disrupted developmental trajectory of prefrontal microcircuitry could lead to altered E/I balance and subsequent deficits in the social domain.
Collapse
Affiliation(s)
- Lucy K Bicks
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Hiroyuki Koike
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA ; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| |
Collapse
|
11
|
Antidepressant/anxiolytic potential and adverse effect liabilities of melanin-concentrating hormone receptor 1 antagonists in animal models. Pharmacol Biochem Behav 2015; 135:154-68. [PMID: 26044968 DOI: 10.1016/j.pbb.2015.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 01/17/2023]
Abstract
Melanin-concentrating hormone receptor 1 (MCH1 receptor) is known to be involved in the control of mood and stress, in addition to the regulation of feeding. Here, we report further evidence that the blockade of the MCH1 receptor exhibits antidepressant and anxiolytic-like effects in a variety of animal models using TASP0382650 and TASP0489838, newly synthesized MCH1 receptor antagonists, with different scaffolds. Both TASP0382650 and TASP0489838 exhibited high affinities for human MCH1 receptor with IC50 values of 7.13 and 3.80nM, respectively. Both compounds showed potent antagonist activities at the MCH1 receptor, as assessed using MCH-increased [(35)S]GTPγS binding to human MCH1 receptor and an MCH-induced [Ca(2+)]i assay in rat MCH1 receptor expressing cells. In contrast, neither TASP0382650 nor TASP0489838 showed an affinity for the MCH2 receptor, another MCH receptor subtype. The oral administration of TASP0382650 or TASP0489838 significantly reduced the immobility time during the forced swimming test in rats, and reduced hyperemotionality induced by an olfactory bulbectomy, both of which are indicative of an antidepressant-like potential. In the olfactory bulbectomy model, the antidepressant effect of TASP0382650 appeared following a single administration, suggesting a faster onset of action, compared with current medications. Moreover, both TASP0382650 and TASP0489838 exhibited anxiolytic effects in several animal models of anxiety. In contrast, both TASP0382650 and TASP0489838 did not affect spontaneous locomotor activity, motor function, spatial memory during the Morris water maze task, or the convulsion threshold to pentylenetetrazole. These findings provide additional evidence that the blockade of the MCH1 receptor exhibits antidepressant- and anxiolytic activities with no adverse effects in experimental animal models.
Collapse
|
12
|
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
|
13
|
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
|
14
|
Testing social acoustic memory in rats: effects of stimulus configuration and long-term memory on the induction of social approach behavior by appetitive 50-kHz ultrasonic vocalizations. Neurobiol Learn Mem 2012; 98:154-64. [PMID: 22677211 DOI: 10.1016/j.nlm.2012.05.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 05/18/2012] [Accepted: 05/23/2012] [Indexed: 02/03/2023]
Abstract
Rats emit distinct types of ultrasonic vocalizations (USVs), which serve as situation-dependent affective signals. In appetitive situations, such as rough-and-tumble-play, high-frequency 50-kHz USVs occur, whereas low-frequency 22-kHz USVs can be observed in aversive situations, such as social defeat. USVs serve distinct communicative functions and induce call-specific behavioral responses in the receiver. While aversive 22-kHz USVs serve as alarm calls and induce behavioral inhibition, appetitive 50-kHz USVs have a pro-social communicative function and elicit social approach behavior, supporting the notion that they serve as social contact calls to (re)establish or maintain contact among conspecifics. The aim of the present study was to use the rat's ability to communicate in the ultrasonic range via high-frequency 50-kHz USVs in order to develop a test for social acoustic memory in rats with relevance for human verbal memory. Verbal learning and memory is among the seven cognitive domains identified as commonly deficient in human schizophrenia patients, but particularly difficult to model. We therefore tested whether the induction of social approach behavior by playback of appetitive 50-kHz USVs is dependent on (1) acoustic stimulus configuration and (2) social long-term memory, and whether (3) social long-term memory effects can be blocked by the administration of scopolamine, a muscarinic acetylcholine antagonist producing amnesia. Results show that social approach behavior in response to playback of natural 50-kHz USVs depends on acoustic stimulus configuration and occurs only when sound energy is concentrated to a critical frequency band in the ultrasonic range. Social approach behavior was detected during the first exposure to playback of 50-kHz USVs, whereas no such response was observed during the second exposure 1week later, indicating a stable memory trace. In contrast, when memory formation was blocked by i.p. administration of scopolamine (0.5mg/kg or 1.5mg/kg) immediately after the first exposure, rats displayed social approach behavior during the second exposure as well. Induction of social approach behavior in response to repeated playback of natural 50-kHz USVs may therefore provide a new and rather unique approach for testing social acoustic memory in rats with relevance to human verbal memory.
Collapse
|
15
|
Fornaro M, Prestia D, Colicchio S, Perugi G. A systematic, updated review on the antidepressant agomelatine focusing on its melatonergic modulation. Curr Neuropharmacol 2011; 8:287-304. [PMID: 21358978 PMCID: PMC3001221 DOI: 10.2174/157015910792246227] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 04/01/2010] [Accepted: 04/08/2010] [Indexed: 12/12/2022] Open
Abstract
Objective: To present an updated, comprehensive review on clinical and pre-clinical studies on agomelatine. Method: A MEDLINE, Psycinfo and Web of Science search (1966-May 2009) was performed using the following keywords: agomelatine, melatonin, S20098, efficacy, safety, adverse effect, pharmacokinetic, pharmacodynamic, major depressive disorder, bipolar disorder, Seasonal Affective Disorder (SAD), Alzheimer, ADHD, Generalized Anxiety Disorder (GAD), Panic Disorder (PD), Obsessive-Compulsive Disorder (OCD), anxiety disorders and mood disorder. Study collection and data extraction: All articles in English identified by the data sources were evaluated. Randomized, controlled clinical trials involving humans were prioritized in the review. The physiological bases of melatonergic transmission were also examined to deepen the clinical comprehension of agomelatine’ melatonergic modulation. Data synthesis: Agomelatine, a melatonergic analogue drug acting as MT1/MT2 agonist and 5-HT2C antagonist, has been reported to be an effective antidepressant therapy. Conclusions: Although a bias in properly assessing the “sleep core” of depression may still exist with current screening instruments, therefore making difficult to compare agomelatine’ efficacy to other antidepressant ones, comparative studies showed agomelatine to be an intriguing option for depression and, potentially, for other therapeutic targets as well.
Collapse
Affiliation(s)
- Michele Fornaro
- Department of Psychiatry, University of Genova, Genoa, Italy
| | | | | | | |
Collapse
|
16
|
Lagos P, Urbanavicius J, Scorza MC, Miraballes R, Torterolo P. Depressive-like profile induced by MCH microinjections into the dorsal raphe nucleus evaluated in the forced swim test. Behav Brain Res 2011; 218:259-66. [DOI: 10.1016/j.bbr.2010.10.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 10/13/2010] [Accepted: 10/22/2010] [Indexed: 10/18/2022]
|
17
|
Chung S, Parks GS, Lee C, Civelli O. Recent updates on the melanin-concentrating hormone (MCH) and its receptor system: lessons from MCH1R antagonists. J Mol Neurosci 2010; 43:115-21. [PMID: 20582487 PMCID: PMC3018593 DOI: 10.1007/s12031-010-9411-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 06/11/2010] [Indexed: 12/01/2022]
Abstract
Melanin-concentrating hormone (MCH) is a 19-amino-acid cyclic peptide which was originally found to lighten skin color in fish that is highly conserved among many species. MCH interacts with two G-protein-coupled receptors, MCH1R and MCH2R, but only MCH1R is expressed in rodents. MCH is mainly synthesized in the lateral hypothalamus and zona incerta, while MCH1R is widely expressed throughout the brain. Thus, MCH signaling is implicated in the regulation of many physiological functions. The identification of MCH1R has led to the development of small-molecule MCH1R antagonists that can block MCH signaling. MCH1R antagonists are useful not only for their potential therapeutic value, but also for understanding the physiological functions of the endogenous MCH system. Here, we review the physiological functions of the MCH system which have been investigated using MCH1R antagonists such as food intake, anxiety, depression, reward, and sleep. This will help us understand the physiological functions of the MCH system and suggest some of the potential applications of MCH1R antagonists in human disorders.
Collapse
Affiliation(s)
- Shinjae Chung
- Department of Pharmacology, University of California, 369 Med Surge II, Irvine, CA 92612, USA
| | | | | | | |
Collapse
|
18
|
Alldredge B. Pathogenic involvement of neuropeptides in anxiety and depression. Neuropeptides 2010; 44:215-24. [PMID: 20096456 DOI: 10.1016/j.npep.2009.12.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 11/15/2009] [Accepted: 12/15/2009] [Indexed: 12/11/2022]
Abstract
Anxiety and depression are highly prevalent disorders of mood posing significant challenges to individuals and society. Current evidence indicates no single neurobiological determinant underpins these conditions and an integrated approach in both research and treatment is expedient. Basic, behavioral, and clinical science indicates various stress-responsive neuropeptides in the neuroendocrine, autonomic, and behavioral pathophysiology of stress-related disorders including anxiety and depression. This review draws on recent research to capture the consensus and implications of neuropeptide research concerning the pathogenesis of anxiety and depression.
Collapse
Affiliation(s)
- Brett Alldredge
- Kansas City University of Medicine and Bioscience, College of Medicine, 1705 Independence Ave., Kansas City, United States.
| |
Collapse
|
19
|
Adamantidis A, de Lecea L. A role for Melanin-Concentrating Hormone in learning and memory. Peptides 2009; 30:2066-70. [PMID: 19576257 PMCID: PMC4287368 DOI: 10.1016/j.peptides.2009.06.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 06/19/2009] [Accepted: 06/22/2009] [Indexed: 11/26/2022]
Abstract
The neurobiological substrate of learning process and persistent memory storage involves multiple brain areas. The neocortex and hippocampal formation are known as processing and storage sites for explicit memory, whereas the striatum, amygdala, neocortex and cerebellum support implicit memory. Synaptic plasticity, long-term changes in synaptic transmission efficacy and transient recruitment of intracellular signaling pathways in these brain areas have been proposed as possible mechanisms underlying short- and long-term memory retention. In addition to the classical neurotransmitters (glutamate, GABA), experimental evidence supports a role for neuropeptides in modulating memory processes. This review focuses on the role of the Melanin-Concentrating Hormone (MCH) and receptors on memory formation in animal studies. Possible mechanisms may involve direct MCH modulation of neural circuit activity that support memory storage and cognitive functions, as well as indirect effect on arousal.
Collapse
Affiliation(s)
- Antoine Adamantidis
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304-5742, USA.
| | | |
Collapse
|
20
|
The Role of Melanin-Concentrating Hormone in Energy Homeostasis and Mood Disorders. J Mol Neurosci 2009; 39:86-98. [DOI: 10.1007/s12031-009-9207-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 04/19/2009] [Indexed: 12/24/2022]
|
21
|
Millan MJ. Dual- and triple-acting agents for treating core and co-morbid symptoms of major depression: novel concepts, new drugs. Neurotherapeutics 2009; 6:53-77. [PMID: 19110199 PMCID: PMC5084256 DOI: 10.1016/j.nurt.2008.10.039] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The past decade of efforts to find improved treatment for major depression has been dominated by genome-driven programs of rational drug discovery directed toward highly selective ligands for nonmonoaminergic agents. Selective drugs may prove beneficial for specific symptoms, for certain patient subpopulations, or both. However, network analyses of the brain and its dysfunction suggest that agents with multiple and complementary modes of action are more likely to show broad-based efficacy against core and comorbid symptoms of depression. Strategies for improved multitarget exploitation of monoaminergic mechanisms include triple inhibitors of dopamine, serotonin (5-HT) and noradrenaline reuptake, and drugs interfering with feedback actions of monoamines at inhibitory 5-HT(1A), 5-HT(1B) and possibly 5-HT(5A) and 5-HT(7) receptors. Specific subsets of postsynaptic 5-HT receptors mediating antidepressant actions are under study (e.g., 5-HT(4) and 5-HT(6)). Association of a clinically characterized antidepressant mechanism with a nonmonoaminergic component of activity is an attractive strategy. For example, agomelatine (a melatonin agonist/5-HT(2C) antagonist) has clinically proven activity in major depression. Dual neurokinin(1) antagonists/5-HT reuptake inhibitors (SRIs) and melanocortin(4) antagonists/SRIs should display advantages over their selective counterparts, and histamine H(3) antagonists/SRIs, GABA(B) antagonists/SRIs, glutamatergic/SRIs, and cholinergic agents/SRIs may counter the compromised cognitive function of depression. Finally, drugs that suppress 5-HT reuptake and blunt hypothalamo-pituitary-adrenocorticotrophic axis overdrive, or that act at intracellular proteins such as GSK-3beta, may abrogate the negative effects of chronic stress on mood and neuronal integrity. This review discusses the discovery and development of dual- and triple-acting antidepressants, focusing on novel concepts and new drugs disclosed over the last 2 to 3 years.
Collapse
Affiliation(s)
- Mark J Millan
- Psychopharmacology Department, Institut du Recherches Servier, Centre de Recherches de Croissy, Paris, France.
| |
Collapse
|
22
|
Nagasaki H, Chung S, Dooley CT, Wang Z, Li C, Saito Y, Clark SD, Houghten RA, Civelli O. The pharmacological properties of a novel MCH1 receptor antagonist isolated from combinatorial libraries. Eur J Pharmacol 2008; 602:194-202. [PMID: 19041642 DOI: 10.1016/j.ejphar.2008.10.068] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 10/09/2008] [Accepted: 10/31/2008] [Indexed: 01/16/2023]
Abstract
Melanin-concentrating hormone (MCH) is a neuropeptide that exhibits potent orexigenic activity. In rodents, it exerts its actions by interacting with one receptor, MCH(1) receptor which is expressed in many parts of the central nervous system (CNS). To study the physiological implications of the MCH system, we need to be able to block it locally and acutely. This necessitates the use of MCH(1) receptor antagonists. While MCH(1) receptor antagonists have been previously reported, they are mainly not accessible to academic research. We apply here a strategy that leads to the isolation of a high affinity and selective MCH(1) receptor antagonist amenable to in vivo analyses without further chemical modifications. This antagonist, TPI 1361-17, was identified through the screening of multiple non-peptide positional scanning synthetic combinatorial libraries (PS-SCL) totaling more than eight hundred thousand compounds in conditions that allow for the identification of only high-affinity compounds. TPI 1361-17 exhibited an IC(50) value of 6.1 nM for inhibition of 1 nM MCH-induced Ca(2+) mobilization and completely displaced the binding of [(125)I] MCH to rat MCH(1) receptor. TPI 1361-17 was found specific, having no affinity for a variety of other G-protein coupled receptors and channels. TPI 1361-17 was found active in vivo since it blocked MCH-induced food intake by 75%. Our results indicate that TPI 1361-17 is a novel and selective MCH(1) receptor antagonist and is an effective tool to study the physiological functions of the MCH system. These results also illustrate the successful application of combinatorial library screening to identify specific surrogate antagonists in an academic setting.
Collapse
Affiliation(s)
- Hiroshi Nagasaki
- Dept. of Metabolic Medicine, School of Medicine, Nagoya University, Nagoya, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|