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 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
|
Zhang Q, Liu X, Ma Q, Zhang J. Melanin concentrating hormone regulates the JNK/ERK signaling pathway to alleviate influenza A virus infection-induced neuroinflammation. J Neuroinflammation 2024; 21:259. [PMID: 39390522 PMCID: PMC11468281 DOI: 10.1186/s12974-024-03251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
Melanin concentrating hormone (MCH) controls many brain functions, such as sleep/wake cycle and memory, and modulates the inflammation response. Previous studies have shown that influenza A virus (IAV) infection-induced neuroinflammation leads to central nervous damage. This study investigated the potential effects of MCH against neuroinflammation induced by IAV infection and its mechanism. MCH (1 and 2 mg/ml) was administrated for 5 consecutive days before IAV infection. Pentobarbital-induced sleep tests, an open-field test, and a Morris water maze were performed to measure sleep quality, spatial learning and memory ability. Neuronal loss and microglial activation were observed with Nissl staining and immunofluorescence assay. The levels of inflammatory cytokines and the expression of the JNK/ERK signaling pathway were examined by ELISA and western blot. IAV infection led to poor sleep quality, impaired the ability of spatial learning and memory, caused neuronal loss and microglial activation in mice's hippocampus and cortex. Meanwhile the level of inflammatory cytokines increased, and the JNK/ERK signaling pathway was activated after IAV infection. MCH administration significantly alleviated IAV-induced neuroinflammation, cognitive impairment, and sleep disorder, decreased the levels of inflammatory cytokines, and inhibited neuronal loss and microglial activation in the hippocampus and cortex by regulating the JNK/ERK signaling pathway. Therefore, MCH alleviated the neuroinflammation, spatial learning and memory impairment, and sleep disorder in IAV-infected mice by regulating the JNK/ERK signaling pathway.
Collapse
Affiliation(s)
- Qianlin Zhang
- Neurology Department, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan Province, 450003, China
| | - Xiaoyang Liu
- Neurology Department, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan Province, 450003, China
| | - Qiankun Ma
- Neurology Department, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan Province, 450003, China
| | - Jiewen Zhang
- Neurology Department, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan Province, 450003, China.
| |
Collapse
|
3
|
Galiautdinova A, Dolgopolova I, Troshina D, Petelin D, Volel B. The Association of Genetic Polymorphisms and Atypical Depression in Adults: A Systematic Review. CLINICAL NEUROPSYCHIATRY 2024; 21:376-384. [PMID: 39540076 PMCID: PMC11555661 DOI: 10.36131/cnfioritieditore20240503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Objective Atypical depression (AD) is a clinical subtype of depression characterised by mood reactivity and at least two of the following features: significant weight gain/increased appetite, hypersomnia, leaden paralysis, and/or interpersonal rejection sensitivity. The role of genetics in the development of depression remains a considerable level of interest among individuals. Due to the large number of breakthrough studies in genetics, there is currently a wealth of heterogeneous data on the existence of genetic markers for depression, including AD. However, it appears that there is a gap in the literature, as we were unable to identify any systematic reviews or meta-analyses that comprehensively describe these data. Therefore, our research aims to provide high-quality, solid evidence for further studies in this area. Method Electronic bibliographic databases (Scopus, MEDLINE) were systematically searched from inception to September 2023. We searched for any specific genetic markers that could be retrieved associated with AD. The quality of studies has been assessed by means of the Q-genie tool. Results Nine studies meeting the inclusion criteria were selected, which appeared to link genetic polymorphisms to atypical depression. Four studies examined genetic polymorphisms associated with the serotonin transporter gene (5-HTT), three studies examined genetic polymorphisms associated with endocrine regulation, two studies considered genetic polymorphisms associated with immune and/or cellular regulation, specifically the melanin-concentrating hormone receptor 2 (MCHR2), mineralocorticoid receptor (MR), and fat mass and obesity-associated protein (FTO) genes involved in the regulation of energy balance. Conclusions The extracted data confirm that the atypical type of major depressive disorder is heritable to a certain extent. Individual risk markers for developing this type of depression may be identified in the future.
Collapse
Affiliation(s)
- Aysylu Galiautdinova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - Iuliia Dolgopolova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - Daria Troshina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - Dmitry Petelin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - Beatrice Volel
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| |
Collapse
|
4
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
5
|
Li XY, Zhang SY, Hong YZ, Chen ZG, Long Y, Yuan DH, Zhao JJ, Tang SS, Wang H, Hong H. TGR5-mediated lateral hypothalamus-dCA3-dorsolateral septum circuit regulates depressive-like behavior in male mice. Neuron 2024; 112:1795-1814.e10. [PMID: 38518778 DOI: 10.1016/j.neuron.2024.02.019] [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: 07/17/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/24/2024]
Abstract
Although bile acids play a notable role in depression, the pathological significance of the bile acid TGR5 membrane-type receptor in this disorder remains elusive. Using depression models of chronic social defeat stress and chronic restraint stress in male mice, we found that TGR5 in the lateral hypothalamic area (LHA) predominantly decreased in GABAergic neurons, the excitability of which increased in depressive-like mice. Upregulation of TGR5 or inhibition of GABAergic excitability in LHA markedly alleviated depressive-like behavior, whereas down-regulation of TGR5 or enhancement of GABAergic excitability facilitated stress-induced depressive-like behavior. TGR5 also bidirectionally regulated excitability of LHA GABAergic neurons via extracellular regulated protein kinases-dependent Kv4.2 channels. Notably, LHA GABAergic neurons specifically innervated dorsal CA3 (dCA3) CaMKIIα neurons for mediation of depressive-like behavior. LHA GABAergic TGR5 exerted antidepressant-like effects by disinhibiting dCA3 CaMKIIα neurons projecting to the dorsolateral septum (DLS). These findings advance our understanding of TGR5 and the LHAGABA→dCA3CaMKIIα→DLSGABA circuit for the development of potential therapeutic strategies in depression.
Collapse
Affiliation(s)
- Xu-Yi Li
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi-Ya Zhang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi-Zhou Hong
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhi-Gang Chen
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Long
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Dan-Hua Yuan
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Jia Zhao
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Su-Su Tang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Wang
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine/Nanhu Brain-Computer Interface Institute, Hangzhou 310013, China.
| | - Hao Hong
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Prochazkova P, Sonka K, Roubalova R, Jezkova J, Nevsimalova S, Buskova J, Merkova R, Dvorakova T, Prihodova I, Dostalova S, Tlaskalova-Hogenova H. Investigation of anti-neuronal antibodies and disparity in central hypersomnias. Sleep Med 2024; 113:220-231. [PMID: 38056084 DOI: 10.1016/j.sleep.2023.11.039] [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: 09/29/2023] [Revised: 11/06/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
STUDY OBJECTIVES Microbial antigens can elicit an immune response leading to the production of autoantibodies cross-reacting with autoantigens. Still, their clinical significance in human sera in the context of brain diseases is unclear. Therefore, assessment of natural autoantibodies reacting with their neuropeptides may elucidate the autoimmune etiology of central hypersomnias. The study aims to determine whether serum autoantibody levels differ in patients with different types of central hypersomnias (narcolepsy type 1 and 2, NT1 and NT2; idiopathic hypersomnia, IH) and healthy controls and if the differences could suggest the participation of autoantibodies in disease pathogenesis. METHODS Sera from 91 patients with NT1, 27 with NT2, 46 with IH, and 50 healthy controls were examined for autoantibodies against assorted neuropeptides. Participants were screened using questionnaires related to sleep disorders, quality of life, and mental health conditions. In addition, serum biochemical parameters and biomarkers of microbial penetration through the intestinal wall were determined. RESULTS A higher prevalence of autoantibodies against neuropeptides was observed only for alpha-melanocytes-stimulating hormone (α-MSH) and neuropeptide glutamic acid-isoleucine (NEI), which differed slightly among diagnoses. Patients with both types of narcolepsy exhibited signs of microbial translocation through the gut barrier. According to the questionnaires, patients diagnosed with NT2 or IH had subjectively worse life quality than patients with NT1. Patients displayed significantly lower levels of bilirubin and creatinine and slightly higher alkaline phosphatase values than healthy controls. CONCLUSIONS Overall, serum anti-neuronal antibodies prevalence is rare, suggesting that their participation in the pathophysiology of concerned sleep disorders is insignificant. Moreover, their levels vary slightly between diagnoses indicating no major diagnostic significance.
Collapse
Affiliation(s)
- Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Janet Jezkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Sona Nevsimalova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Jitka Buskova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radana Merkova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tereza Dvorakova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Iva Prihodova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Simona Dostalova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
9
|
Kurban N, Qin Y, Zhao HL, Hu X, Chen X, Zhao YY, Peng YS, Wang HB, Cui SY, Zhang YH. Chronic Stress-Induced Elevation of Melanin-Concentrating Hormone in the Locus Coeruleus Inhibits Norepinephrine Production and Associated With Depression-Like Behaviors in Rats. Int J Neuropsychopharmacol 2024; 27:pyad069. [PMID: 38135278 PMCID: PMC10799331 DOI: 10.1093/ijnp/pyad069] [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: 01/21/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide that projects throughout the central nervous system, including the noradrenergic locus coeruleus (LC). Our previous study suggested that MCH/MCH receptor 1 (MCHR1) in the LC may be involved in the regulation of depression. The present study investigated whether the role of MCH/MCHR1 in the LC in depression-like behaviors is associated with the regulation of norepinephrine. METHOD Chronic unpredictable stress (CUS) and an acute intra-LC microinjection of MCH induced depression-like behaviors in rats. The MCHR1 antagonist SNAP-94847 was also microinjected in the LC in rats that were suffering CUS or treated with MCH. The sucrose preference, forced swim, and locomotor tests were used for behavioral evaluation. Immunofluorescence staining, enzyme-linked immunosorbent assay, western blot, and high-performance liquid chromatography with electrochemical detection were used to explore the mechanism of MCH/MCHR1 in the regulation of depression-like behaviors. RESULTS CUS induced an abnormal elevation of MCH levels and downregulated MCHR1 in the LC, which was highly correlated with the formation of depression-like behaviors. SNAP-94847 exerted antidepressant effects in CUS-exposed rats by normalizing tyrosine hydroxylase, dopamine β hydroxylase, and norepinephrine in the LC. An acute microinjection of MCH induced depression-like behaviors through its action on MCHR1. MCHR1 antagonism in the LC significantly reversed the MCH-induced downregulation of norepinephrine production by normalizing MCHR1-medicated cAMP-PKA signaling. CONCLUSIONS Our study confirmed that the MCH/MCHR1 system in the LC may be involved in depression-like behaviors by downregulating norepinephrine production. These results improve our understanding of the pathogenesis of depression that is related to the MCH/MCHR1 system in the LC.
Collapse
Affiliation(s)
- Nurhumar Kurban
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Yu Qin
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Hui-Ling Zhao
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Xiao Hu
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Xi Chen
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Yi-Yi Zhao
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Yu-Shuo Peng
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| | - Hong-Bo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai, China
| | - Su-Ying Cui
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai, China
| | - Yong-He Zhang
- Department of Pharmacology, Peking University, School of Basic Medical Science, Beijing, China
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
Alcantara-Zapata DE, Lucero N, De Gregorio N, Astudillo Cornejo P, Ibarra Villanueva C, Baltodano-Calle MJ, Gonzales GF, Behn C. Women's mood at high altitude. sexual dimorphism in hypoxic stress modulation by the tryptophan-melatonin axis. Front Physiol 2023; 13:1099276. [PMID: 36733695 PMCID: PMC9887123 DOI: 10.3389/fphys.2022.1099276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Sexual (and gender)-dimorphism in tolerance to hypobaric hypoxia increasingly matters for a differential surveillance of human activities at high altitude (HA). At low altitudes, the prevalence of anxiety and depression in women has already been found to double when compared with men; it could be expected to even increase on exposure to HA. In purposefully caring for the health of women at HA, the present work explores the potential involvement of the tryptophan (Trp)-melatonin axis in mood changes on exposure to hypobaric hypoxia. The present work highlights some already known anxiogenic effects of HA exposure. Hypoxia and insomnia reduce serotonin (5-HT) availability; the latter defect being expressed as failure of brown adipose tissue (BAT) activation and mood disorders. Rapid eye movement (REM) sleep organization and synapsis restoration that are additionally affected by hypoxia impair memory consolidation. Affective complaints may thus surge, evolving into anxiety and depression. Sex-related differences in neural network organization and hormonal changes during the menstrual cycle, and certainly also during the life cycle, underscore the possibility of 5-HT-related mood alterations, particularly in women on HA exposure. The mean brain rate of 5-HT synthesis at sea level is already 1.5-fold higher in males than in females. sexual dimorphism also evidences the overexpression effects of SERT, a 5-HT transporter protein. Gonadal and thyroid hormones, as influenced by HA exposure, further modulate 5-HT availability and its effects in women. Besides caring for adequate oxygenation and maintenance of one's body core temperature, special precautions concerning women sojourning at HA should include close observations of hormonal cycles and, perhaps, also trials with targeted antidepressants.
Collapse
Affiliation(s)
- D. E. Alcantara-Zapata
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo (LID), Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - N. Lucero
- Occupational Health Program, School of Public Health, University of Chile, Santiago, Chile
| | - N. De Gregorio
- Laboratory of Extreme Environments, Department of Physiology and Biophysics, Biomedical Science Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - P. Astudillo Cornejo
- Occupational Ergonomics Program, Department of Kinesiology, University of Atacama, Copiapó, Chile
| | - C. Ibarra Villanueva
- Occupational Ergonomics Program, Department of Kinesiology, University of Atacama, Copiapó, Chile
| | - M. J. Baltodano-Calle
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo (LID), Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - G. F. Gonzales
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo (LID), Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
- High Altitude Research Institute, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - C. Behn
- Laboratory of Extreme Environments, Department of Physiology and Biophysics, Biomedical Science Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Faculty of Medicine, University of Atacama, Copiapó, Chile
| |
Collapse
|
12
|
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: 0.5] [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
|
13
|
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: 1.5] [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
|
14
|
He X, Li Y, Zhang N, Huang J, Ming X, Guo R, Hu Y, Ji P, Guo F. Melanin-concentrating hormone promotes anxiety and intestinal dysfunction via basolateral amygdala in mice. Front Pharmacol 2022; 13:906057. [PMID: 36016574 PMCID: PMC9395614 DOI: 10.3389/fphar.2022.906057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
The limbic system plays a pivotal role in stress-induced anxiety and intestinal disorders, but how the functional circuits between nuclei within the limbic system are engaged in the processing is still unclear. In our study, the results of fluorescence gold retrograde tracing and fluorescence immunohistochemistry showed that the melanin-concentrating hormone (MCH) neurons of the lateral hypothalamic area (LHA) projected to the basolateral amygdala (BLA). Both chemogenetic activation of MCH neurons and microinjection of MCH into the BLA induced anxiety disorder in mice, which were reversed by intra-BLA microinjection of MCH receptor 1 (MCHR1) blocker SNAP-94847. In the chronic acute combining stress (CACS) stimulated mice, SNAP94847 administrated in the BLA ameliorated anxiety-like behaviors and improved intestinal dysfunction via reducing intestinal permeability and inflammation. In conclusion, MCHergic circuit from the LHA to the BLA participates in the regulation of anxiety-like behavior in mice, and this neural pathway is related to the intestinal dysfunction in CACS mice by regulating intestinal permeability and inflammation.
Collapse
Affiliation(s)
- Xiaoman He
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yuhang Li
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Nana Zhang
- Department of Clinical Laboratory, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinfang Huang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xing Ming
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ruixiao Guo
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yang Hu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Pengfei Ji
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Feifei Guo
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, China
- *Correspondence: Feifei Guo,
| |
Collapse
|
15
|
Rodrigues LTC, Patrone LGA, Gargaglioni LH, Dias MB. Melanin-concentrating hormone regulates the hypercapnic chemoreflex by acting in the lateral hypothalamic area. Exp Physiol 2022; 107:1298-1311. [PMID: 35930596 DOI: 10.1113/ep090318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? MCH suppresses the hypercapnic chemoreflex but the mechanism by which this effect is produced has not been previously explored. What is the main finding and its importance? MCH acting in the lateral hypothalamic area but not in the locus coeruleus in rats, in the light period, attenuates the hypercapnic chemoreflex. Our data provide new insight regarding the role of MCH in the modulation of the hypercapnic ventilatory response. ABSTRACT Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide involved in a broad range of homeostatic functions including regulation of the hypercapnic chemoreflex. We evaluated whether MCH modulates the hypercapnic ventilatory response by acting in the lateral hypothalamic area (LHA) and/or in the locus coeruleus (LC). Here, we measured pulmonary ventilation (VE ), body temperature, electroencephalogram (EEG) and electromyogram (EMG) of unanesthetized adult male Wistar rats before and after microinjection of MCH [0.4 mM] or MCH1-R antagonist (SNAP-94847 [63 mM]) into the LHA and LC, in room air and 7% CO2 conditions during wakefulness and sleep, in the dark and light periods. MCH intra-LHA caused a decreased CO2 ventilatory response during wakefulness and sleep in the light period, while SNAP-94847 intra-LHA increased this response, during wakefulness in the light period. In the LC, MCH or the MCH1-R antagonist caused no change in the hypercapnic ventilatory response. Our results suggest that MCH, in the LHA, exerts an inhibitory modulation of the hypercapnic ventilatory response during the light-inactive period in rats. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laísa T C Rodrigues
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Mirela B Dias
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| |
Collapse
|
16
|
Cocaine-induced neural adaptations in the lateral hypothalamic melanin-concentrating hormone neurons and the role in regulating rapid eye movement sleep after withdrawal. Mol Psychiatry 2021; 26:3152-3168. [PMID: 33093653 PMCID: PMC8060355 DOI: 10.1038/s41380-020-00921-1] [Citation(s) in RCA: 5] [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: 05/20/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022]
Abstract
Sleep abnormalities are often a prominent contributor to withdrawal symptoms following chronic drug use. Notably, rapid eye movement (REM) sleep regulates emotional memory, and persistent REM sleep impairment after cocaine withdrawal negatively impacts relapse-like behaviors in rats. However, it is not understood how cocaine experience may alter REM sleep regulatory machinery, and what may serve to improve REM sleep after withdrawal. Here, we focus on the melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus (LH), which regulate REM sleep initiation and maintenance. Using adult male Sprague-Dawley rats trained to self-administer intravenous cocaine, we did transcriptome profiling of LH MCH neurons after long-term withdrawal using RNA-sequencing, and performed functional assessment using slice electrophysiology. We found that 3 weeks after withdrawal from cocaine, LH MCH neurons exhibit a wide range of gene expression changes tapping into cell membrane signaling, intracellular signaling, and transcriptional regulations. Functionally, they show reduced membrane excitability and decreased glutamatergic receptor activity, consistent with increased expression of voltage-gated potassium channel gene Kcna1 and decreased expression of metabotropic glutamate receptor gene Grm5. Finally, chemogenetic or optogenetic stimulations of LH MCH neural activity increase REM sleep after long-term withdrawal with important differences. Whereas chemogenetic stimulation promotes both wakefulness and REM sleep, optogenetic stimulation of these neurons in sleep selectively promotes REM sleep. In summary, cocaine exposure persistently alters gene expression profiles and electrophysiological properties of LH MCH neurons. Counteracting cocaine-induced hypoactivity of these neurons selectively in sleep enhances REM sleep quality and quantity after long-term withdrawal.
Collapse
|
17
|
Abstract
Sleep and wakefulness are complex, tightly regulated behaviors that occur in virtually all animals. With recent exciting developments in neuroscience methodologies such as optogenetics, chemogenetics, and cell-specific calcium imaging technology, researchers can advance our understanding of how discrete neuronal groups precisely modulate states of sleep and wakefulness. In this chapter, we provide an overview of key neurotransmitter systems, neurons, and circuits that regulate states of sleep and wakefulness. We also describe long-standing models for the regulation of sleep/wake and non-rapid eye movement/rapid eye movement cycling. We contrast previous knowledge derived from classic approaches such as brain stimulation, lesions, cFos expression, and single-unit recordings, with emerging data using the newest technologies. Our understanding of neural circuits underlying the regulation of sleep and wakefulness is rapidly evolving, and this knowledge is critical for our field to elucidate the enigmatic function(s) of sleep.
Collapse
|
18
|
Pascovich C, Niño S, Mondino A, Lopez-Hill X, Urbanavicius J, Monti J, Lagos P, Torterolo P. Microinjection of melanin-concentrating hormone (MCH) into the median raphe nucleus promotes REM sleep in rats. Sleep Sci 2021; 14:229-235. [PMID: 35186201 PMCID: PMC8848522 DOI: 10.5935/1984-0063.20200075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/30/2020] [Indexed: 11/20/2022] Open
Abstract
Melanin concentrating hormone (MCH) is a sleep-promoting neuromodulator synthesized by neurons located in the postero-lateral hypothalamus and incerto-hypothalamic area. MCHergic neurons have widespread projections including the serotonergic dorsal (DR) and median (MnR) raphe nuclei, both involved in the control of wakefulness and sleep. In the present study, we explored in rats the presence of the MCH receptor type 1 (MCHR-1) in serotonergic neurons of the MnR by double immunofluorescence. Additionally, we analyzed the effect on sleep of MCH microinjections into the MnR. We found that MCHR-1 protein was present in MnR serotonergic and non-serotonergic neurons. In this respect, the receptor was localized in the primary cilia of these neurons. Compared with saline, microinjections of MCH into the MnR induced a dose-related increase in REM sleep time, which was related to a rise in the number of REM sleep episodes, associated with a reduction in the time spent in W. No significant changes were observed in non-REM (NREM) sleep time. Our data strongly suggest that MCH projections towards the MnR, acting through the MCHR-1 located in the primary cilia, promote REM sleep.
Collapse
Affiliation(s)
- Claudia Pascovich
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Sofia Niño
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Alejandra Mondino
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Ximena Lopez-Hill
- Instituto de Investigaciones Biológicas Clemente Estable, Neurofarmacología Experimental, Montevideo - Uruguay
| | - Jessika Urbanavicius
- Instituto de Investigaciones Biológicas Clemente Estable, Neurofarmacología Experimental, Montevideo - Uruguay
| | - Jaime Monti
- Hospital de Clínicas, Farmacología y Terapéutica, Montevideo - Uruguay
| | - Patricia Lagos
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Pablo Torterolo
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay. ,Corresponding author: Pablo Torterolo. E-mail: /
| |
Collapse
|
19
|
Oh JY, Liu QF, Hua C, Jeong HJ, Jang JH, Jeon S, Park HJ. Intranasal Administration of Melanin-Concentrating Hormone Reduces Stress-Induced Anxiety- and Depressive-Like Behaviors in Rodents. Exp Neurobiol 2020; 29:453-469. [PMID: 33372169 PMCID: PMC7788308 DOI: 10.5607/en20024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Major depressive disorder is a complex neuropsychiatric disorder with few treatment options. Non-targeted antidepressants have low efficacy and can induce series of side effects. While a neuropeptide, melanin-concentrating hormone (MCH), is known to exhibit regulator of affective state, no study to date has assessed the anti-depressive effects of MCH in a stress-induced depression model. This study aimed to evaluate the pharmacological effects of intranasal administration of MCH on depression-related behavior in stressed rats and mice. Using a number of behavioral tests, we found that MCH treatment significantly decreased anxiety- and depressive-like behaviors induced by stress. Notably, the effects of MCH were equivalent to those of fluoxetine. MCH treatment also restored the activity of the mammalian target of rapamycin (mTOR) signaling pathway and normalized the levels of synaptic proteins, including postsynaptic density 95, glutamate receptor 1, and synapsin 1, which were all downregulated by stress. Interestingly, the protective effects of MCH were blocked by the mTOR inhibitor, rapamycin. These results suggest that MCH exhibits antidepressant properties by modulating the mTOR pathway. Altogether, this study provides an insight into the molecular mechanisms involved in the antidepressant-like effects of MCH, thereby paving the way for the future clinical application of MCH.
Collapse
Affiliation(s)
- Ju-Young Oh
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.,Studies of Translational Acupuncture Research (STAR), Acupuncture & Meridian Science Research Center (AMSRC), Kyung Hee University, Seoul 02447, Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Quan Feng Liu
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gwangju 38066, Korea
| | - Cai Hua
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju 61469, Korea
| | - Ha Jin Jeong
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju 61469, Korea
| | - Jae-Hwan Jang
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.,Studies of Translational Acupuncture Research (STAR), Acupuncture & Meridian Science Research Center (AMSRC), Kyung Hee University, Seoul 02447, Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju 61469, Korea
| | - Hi-Joon Park
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.,Studies of Translational Acupuncture Research (STAR), Acupuncture & Meridian Science Research Center (AMSRC), Kyung Hee University, Seoul 02447, Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
20
|
Neurochemical regulators of food behavior for pharmacological treatment of obesity: current status and future prospects. Future Med Chem 2020; 12:1865-1884. [PMID: 33040605 DOI: 10.4155/fmc-2019-0361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In recent decades, obesity has become a pandemic disease and appears to be an ultimate medical and social problem. Existing antiobesity drugs show low efficiency and a wide variety of side effects. In this review, we discuss possible mechanisms underlying brain-gut-adipose tissue axis, as well as molecular biochemical characteristics of various neurochemical regulators of body weight and appetite. Multiple brain regions are responsible for eating behavior, hedonic eating and food addiction. The existing pharmacological targets for treatment of obesity were reviewed as well.
Collapse
|
21
|
Diniz GB, Battagello DS, Klein MO, Bono BSM, Ferreira JGP, Motta‐Teixeira LC, Duarte JCG, Presse F, Nahon J, Adamantidis A, Chee MJ, Sita LV, Bittencourt JC. Ciliary melanin‐concentrating hormone receptor 1 (MCHR1) is widely distributed in the murine CNS in a sex‐independent manner. J Neurosci Res 2020; 98:2045-2071. [DOI: 10.1002/jnr.24651] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/24/2020] [Accepted: 05/07/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Giovanne B. Diniz
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
- Department of Neurosurgery Yale School of Medicine New Haven CT USA
| | - Daniella S. Battagello
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Marianne O. Klein
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | | | - Jozélia G. P. Ferreira
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Livia C. Motta‐Teixeira
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Jessica C. G. Duarte
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Françoise Presse
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC) Université Côte d’AzurCNRS Valbonne France
| | - Jean‐Louis Nahon
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC) Université Côte d’AzurCNRS Valbonne France
| | | | - Melissa J. Chee
- Department of Neuroscience Carleton University Ottawa ON Canada
| | - Luciane V. Sita
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Jackson C. Bittencourt
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
- Center for Neuroscience and Behavior Institute of Psychology University of Sao Paulo Sao Paulo Brazil
| |
Collapse
|
22
|
Association of incident angina pectoris and rapid eye movement sleep in a large community-based study: the sleep heart health study. Sleep Med 2019; 59:7-14. [DOI: 10.1016/j.sleep.2018.10.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/30/2018] [Accepted: 10/23/2018] [Indexed: 11/19/2022]
|
23
|
Niño-Rivero S, Torterolo P, Lagos P. Melanin-concentrating hormone receptor-1 is located in primary cilia of the dorsal raphe neurons. J Chem Neuroanat 2019; 98:55-62. [DOI: 10.1016/j.jchemneu.2019.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/25/2019] [Accepted: 03/22/2019] [Indexed: 12/19/2022]
|
24
|
Diniz GB, Battagello DS, Cherubini PM, Reyes-Mendoza JD, Luna-Illades C, Klein MO, Motta-Teixeira LC, Sita LV, Miranda-Anaya M, Morales T, Bittencourt JC. Melanin-concentrating hormone peptidergic system: Comparative morphology between muroid species. J Comp Neurol 2019; 527:2973-3001. [PMID: 31152440 DOI: 10.1002/cne.24723] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022]
Abstract
Melanin-concentrating hormone (MCH) is a conserved neuropeptide, predominantly located in the diencephalon of vertebrates, and associated with a wide range of functions. While functional studies have focused on the use of the traditional mouse laboratory model, critical gaps exist in our understanding of the morphology of the MCH system in this species. Even less is known about the nontraditional animal model Neotomodon alstoni (Mexican volcano mouse). A comparative morphological study among these rodents may, therefore, contribute to a better understanding of the evolution of the MCH peptidergic system. To this end, we employed diverse immunohistochemical protocols to identify key aspects of the MCH system, including its spatial relationship to another neurochemical population of the tuberal hypothalamus, the orexins. Three-dimensional (3D) reconstructions were also employed to convey a better sense of spatial distribution to these neurons. Our results show that the distribution of MCH neurons in all rodents studied follows a basic plan, but individual characteristics are found for each species, such as the preeminence of a periventricular group only in the rat, the lack of posterior groups in the mouse, and the extensive presence of MCH neurons in the anterior hypothalamic area of Neotomodon. Taken together, these data suggest a strong anatomical substrate for previously described functions of the MCH system, and that particular neurochemical and morphological features may have been determinant to species-specific phenotypes in rodent evolution.
Collapse
Affiliation(s)
- Giovanne B Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniella S Battagello
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Pedro M Cherubini
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Julio D Reyes-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Cesar Luna-Illades
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Marianne O Klein
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lívia C Motta-Teixeira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luciane V Sita
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Manuel Miranda-Anaya
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Queretaro, Mexico
| | - Jackson C Bittencourt
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Center for Neuroscience and Behavior, Institute of Psychology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Benarroch EE. Control of the cardiovascular and respiratory systems during sleep. Auton Neurosci 2019; 218:54-63. [DOI: 10.1016/j.autneu.2019.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 01/01/2023]
|
26
|
Benarroch EE. Brainstem integration of arousal, sleep, cardiovascular, and respiratory control. Neurology 2018; 91:958-966. [PMID: 30355703 DOI: 10.1212/wnl.0000000000006537] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
27
|
Research into an Association between Anhedonia and Decreased REM Latency in Moderately to Severely Depressed Patients. SLEEP DISORDERS 2018; 2018:1636574. [PMID: 30057824 PMCID: PMC6051023 DOI: 10.1155/2018/1636574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/16/2018] [Indexed: 11/17/2022]
Abstract
Anhedonia stands as a core symptom and potential trait marker of major depressive disorder (MDD). The importance of rapid eye movement sleep latency (REML) as a biological marker of depression has previously and repeatedly been studied. The aim of this paper is to analyse the relationship between anhedonia and REML in moderately to severely depressed patients. The shortened Beck Depression Inventory (BDI-13) was chosen to assess depressive symptoms and, among them, more particularly, anhedonic symptoms. Two-way ANCOVA was used for statistical analyses. A significant association between anhedonic symptoms and REML was found when the number of sleep cycles (NCy) and the severity of depression were added as covariates. Our findings suggest that REML may be a useful variable to differentiate some diagnostic subtypes of depression related to anhedonia.
Collapse
|
28
|
Tesoriero C, Del Gallo F, Bentivoglio M. Sleep and brain infections. Brain Res Bull 2018; 145:59-74. [PMID: 30016726 DOI: 10.1016/j.brainresbull.2018.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022]
Abstract
Sleep is frequently altered in systemic infections as a component of sickness behavior in response to inflammation. Sleepiness in sickness behavior has been extensively investigated. Much less attention has instead been devoted to sleep and wake alterations in brain infections. Most of these, as other neuroinfections, are prevalent in sub-Saharan Africa. The present overview highlights the importance of this topic from both the clinical and pathogenetic points of view. Vigilance states and their regulation are first summarized, emphasizing that key nodes in this distributed brain system can be targeted by neuroinflammatory signaling. Sleep-wake changes in the parasitic disease human African trypanosomiasis (HAT) and its animal models are then reviewed and discussed. Experimental data have revealed that the suprachiasmatic nucleus, the master circadian pacemaker, and peptidergic cell populations of the lateral hypothalamus (the wake-promoting orexin neurons and the sleep-promoting melanin-concentrating hormone neurons) are targeted by African trypanosome infection. It is then discussed how prominent and disturbing are sleep changes in HIV/AIDS, also when the infection is cured with antiretroviral therapy. This recalls attention on the bidirectional interactions between sleep and immune system, including the specialized brain immune response of which microglial cells are protagonists. Sleep changes in an ancient viral disease, rabies, and in the emerging infection due to Zika virus which causes a congenital syndrome, are also dealt with. Altogether the findings indicate that sleep-wake regulation is targeted by brain infections caused by different pathogens and, although the relevant pathogenetic mechanisms largely remain to be clarified, these alterations differ from hypersomnia occurring in sickness behavior. Thus, brain infections point to the vulnerability of the neural network of sleep-wake regulation as a highly relevant clinical and basic science challenge.
Collapse
Affiliation(s)
- Chiara Tesoriero
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Federico Del Gallo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Marina Bentivoglio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy.
| |
Collapse
|
29
|
Riveros ME, Retamal MA. Are Polyunsaturated Fatty Acids Implicated in Histaminergic Dysregulation in Bipolar Disorder?: AN HYPOTHESIS. Front Physiol 2018; 9:693. [PMID: 29946266 PMCID: PMC6005883 DOI: 10.3389/fphys.2018.00693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/18/2018] [Indexed: 12/28/2022] Open
Abstract
Bipolar disorder (BD) is an extremely disabling psychiatric disease, characterized by alternate states of mania (or hypomania) and depression with euthymic states in between. Currently, patients receive pharmacological treatment with mood stabilizers, antipsychotics, and antidepressants. Unfortunately, not all patients respond well to this type of treatment. Bipolar patients are also more prone to heart and metabolic diseases as well as a higher risk of suicide compared to the healthy population. For a correct brain function is indispensable a right protein and lipids (e.g., fatty acids) balance. In particular, the amount of fatty acids in the brain corresponds to a 50–70% of the dry weight. It has been reported that in specific brain regions of BD patients there is a reduction in the content of unsaturated n-3 fatty acids. Accordingly, a diet rich in n-3 fatty acids has beneficial effects in BD patients, while their absence or high levels of saturated fatty acids in the diet are correlated to the risk of developing the disease. On the other hand, the histamine system is likely to be involved in the pathophysiology of several psychiatric diseases such as BD. Histamine is a neuromodulator involved in arousal, motivation, and energy balance; drugs acting on the histamine receptor H3 have shown potential as antidepressants and antipsychotics. The histaminergic system as other neurotransmission systems can be altered by fatty acid membrane composition. The purpose of this review is to explore how polyunsaturated fatty acids content alterations are related to the histaminergic system modulation and their impact in BD pathophysiology.
Collapse
Affiliation(s)
- María E Riveros
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.,Center of Applied Ecology and Sustainability, Santiago, Chile
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.,Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
30
|
Tsuneki H, Wada T, Sasaoka T. Chronopathophysiological implications of orexin in sleep disturbances and lifestyle-related disorders. Pharmacol Ther 2018; 186:25-44. [DOI: 10.1016/j.pharmthera.2017.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
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: 3.3] [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
|
32
|
Laperchia C, Xu YZ, Mumba Ngoyi D, Cotrufo T, Bentivoglio M. Neural Damage in Experimental Trypanosoma brucei gambiense Infection: Hypothalamic Peptidergic Sleep and Wake-Regulatory Neurons. Front Neuroanat 2018. [PMID: 29535612 PMCID: PMC5835115 DOI: 10.3389/fnana.2018.00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuron populations of the lateral hypothalamus which synthesize the orexin (OX)/hypocretin or melanin-concentrating hormone (MCH) peptides play crucial, reciprocal roles in regulating wake stability and sleep. The disease human African trypanosomiasis (HAT), also called sleeping sickness, caused by extracellular Trypanosoma brucei (T. b.) parasites, leads to characteristic sleep-wake cycle disruption and narcoleptic-like alterations of the sleep structure. Previous studies have revealed damage of OX and MCH neurons during systemic infection of laboratory rodents with the non-human pathogenic T. b. brucei subspecies. No information is available, however, on these peptidergic neurons after systemic infection with T. b. gambiense, the etiological agent of 97% of HAT cases. The present study was aimed at the investigation of immunohistochemically characterized OX and MCH neurons after T. b. gambiense or T. b. brucei infection of a susceptible rodent, the multimammate mouse, Mastomysnatalensis. Cell counts and evaluation of OX fiber density were performed at 4 and 8 weeks post-infection, when parasites had entered the brain parenchyma from the periphery. A significant decrease of OX neurons (about 44% reduction) and MCH neurons (about 54% reduction) was found in the lateral hypothalamus and perifornical area at 8 weeks in T. b. gambiense-infected M. natalensis. A moderate decrease (21% and 24% reduction, respectively), which did not reach statistical significance, was found after T. b. brucei infection. In two key targets of diencephalic orexinergic innervation, the peri-suprachiasmatic nucleus (SCN) region and the thalamic paraventricular nucleus (PVT), densitometric analyses showed a significant progressive decrease in the density of orexinergic fibers in both infection paradigms, and especially during T. b. gambiense infection. Altogether the findings provide novel information showing that OX and MCH neurons are highly vulnerable to chronic neuroinflammatory signaling caused by the infection of human-pathogenic African trypanosomes.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Yuan-Zhong Xu
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Dieudonné Mumba Ngoyi
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of Congo
| | - Tiziana Cotrufo
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Bentivoglio
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,National Institute of Neuroscience (INN), Verona Unit, Verona, Italy
| |
Collapse
|
33
|
Novel analgesic effects of melanin-concentrating hormone on persistent neuropathic and inflammatory pain in mice. Sci Rep 2018; 8:707. [PMID: 29335480 PMCID: PMC5768747 DOI: 10.1038/s41598-018-19145-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022] Open
Abstract
The melanin-concentrating hormone (MCH) is a peptidergic neuromodulator synthesized by neurons in the lateral hypothalamus and zona incerta. MCHergic neurons project throughout the central nervous system, indicating the involvements of many physiological functions, but the role in pain has yet to be determined. In this study, we found that pMCH-/- mice showed lower baseline pain thresholds to mechanical and thermal stimuli than did pMCH+/+ mice, and the time to reach the maximum hyperalgesic response was also significantly earlier in both inflammatory and neuropathic pain. To examine its pharmacological properties, MCH was administered intranasally into mice, and results indicated that MCH treatment significantly increased mechanical and thermal pain thresholds in both pain models. Antagonist challenges with naltrexone (opioid receptor antagonist) and AM251 (cannabinoid 1 receptor antagonist) reversed the analgesic effects of MCH in both pain models, suggesting the involvement of opioid and cannabinoid systems. MCH treatment also increased the expression and activation of CB1R in the medial prefrontal cortex and dorsolateral- and ventrolateral periaqueductal grey. The MCH1R antagonist abolished the effects induced by MCH. This is the first study to suggest novel analgesic actions of MCH, which holds great promise for the application of MCH in the therapy of pain-related diseases.
Collapse
|
34
|
Jancsik V, Bene R, Sótonyi P, Zachar G. Sub-cellular organization of the melanin-concentrating hormone neurons in the hypothalamus. Peptides 2018; 99:56-60. [PMID: 29108810 DOI: 10.1016/j.peptides.2017.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/30/2017] [Accepted: 11/02/2017] [Indexed: 12/22/2022]
Abstract
Melanin-concentrating hormone (MCH) is a potent orexigenic and sleep-promoting neuropeptide in mammals produced predominately by hypothalamic neurons which project to a wide variety of brain areas. Several MCH producing neurons contain MCH as the only neuropeptide, while others comprise cocaine- and amphetamine regulated transcript (CART) as well. The intrahypothalamic localization and the projection pattern of these two subpopulations are distinct. To provide structural grounding to understand the mechanism of action of MCH neurons we show here the subcellular localization of the neuropeptides in the two subpopulations within the hypothalamus of healthy young male mice by applying single and double immunofluorescence labelling.; Thick, prominent MCH immunopositive reticulation and fine discrete granules are detected within the perikarya of both CART positive and CART-free MCH neurons. Typically, one or more immunoreactive processes emanate from the perikarya. The bulk of CART immunoreactivity is also centrally positioned, surrounded by sparse immunoreactive granules within the perikarya and in the processes. In double immunopositive neurons, the two neuropeptides seem to colocalize in the heavily labelled central area, while the immunopositive granules in the cell body periphery and in the processes apparently contain either MCH or CART. This spatial arrangement suggests that MCH and CART, after being synthetized and processed in the endoplasmic reticulum/Golgi complex, are sorted into separate dense core vesicles, which then enter into the cell processes. This mechanism allows for both concerted and independent regulation of the transport and release of MCH and CART.
Collapse
Affiliation(s)
- Veronika Jancsik
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary.
| | - Roland Bene
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Péter Sótonyi
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Gergely Zachar
- Department of Anatomy, Histology and Embryology, Semmelweis University Budapest, Hungary
| |
Collapse
|
35
|
An obstructive sleep apnea primer: What the practicing allergist needs to know. Ann Allergy Asthma Immunol 2017; 118:259-268. [PMID: 28284532 DOI: 10.1016/j.anai.2016.07.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022]
|
36
|
Al Shweiki MHDR, Oeckl P, Steinacker P, Hengerer B, Schönfeldt-Lecuona C, Otto M. Major depressive disorder: insight into candidate cerebrospinal fluid protein biomarkers from proteomics studies. Expert Rev Proteomics 2017; 14:499-514. [DOI: 10.1080/14789450.2017.1336435] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - Patrick Oeckl
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Bastian Hengerer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
| |
Collapse
|
37
|
Abstract
While some autoimmune disorders remain extremely rare, others largely predominate the epidemiology of human autoimmunity. Notably, these include psoriasis, diabetes, vitiligo, thyroiditis, rheumatoid arthritis and multiple sclerosis. Thus, despite the quasi-infinite number of "self" antigens that could theoretically trigger autoimmune responses, only a limited set of antigens, referred here as superautoantigens, induce pathogenic adaptive responses. Several lines of evidence reviewed in this paper indicate that, irrespective of the targeted organ (e.g. thyroid, pancreas, joints, brain or skin), a significant proportion of superautoantigens are highly expressed in the synaptic compartment of the central nervous system (CNS). Such an observation applies notably for GAD65, AchR, ribonucleoproteins, heat shock proteins, collagen IV, laminin, tyrosine hydroxylase and the acetylcholinesterase domain of thyroglobulin. It is also argued that cognitive alterations have been described in a number of autoimmune disorders, including psoriasis, rheumatoid arthritis, lupus, Crohn's disease and autoimmune thyroiditis. Finally, the present paper points out that a great majority of the "incidental" autoimmune conditions notably triggered by neoplasms, vaccinations or microbial infections are targeting the synaptic or myelin compartments. On this basis, the concept of an immunological homunculus, proposed by Irun Cohen more than 25 years ago, is extended here in a model where physiological autoimmunity against brain superautoantigens confers both: i) a crucial evolutionary-determined advantage via cognition-promoting autoimmunity; and ii) a major evolutionary-determined vulnerability, leading to the emergence of autoimmune disorders in Homo sapiens. Moreover, in this theoretical framework, the so called co-development/co-evolution model, both the development (at the scale of an individual) and evolution (at the scale of species) of the antibody and T-cell repertoires are coupled to those of the neural repertoires (i.e. the distinct neuronal populations and synaptic circuits supporting cognitive and sensorimotor functions). Clinical implications and future experimental insights are also presented and discussed.
Collapse
Affiliation(s)
- Serge Nataf
- Bank of Tissues and Cells, Lyon University Hospital (Hospices Civils de Lyon), CarMeN Laboratory, INSERM 1060, INRA 1397, INSA Lyon, Université Claude Bernard Lyon-1, Lyon, F-69000, France
| |
Collapse
|
38
|
Van Camp KA, Baggerman G, Blust R, Husson SJ. Peptidomics of the zebrafish Danio rerio : In search for neuropeptides. J Proteomics 2017; 150:290-296. [DOI: 10.1016/j.jprot.2016.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/07/2016] [Accepted: 09/27/2016] [Indexed: 12/27/2022]
|
39
|
Ansari M, Rahmani F, Dolatshahi M, Pooyan A, Aarabi MH. Brain pathway differences between Parkinson’s disease patients with and without REM sleep behavior disorder. Sleep Breath 2016; 21:155-161. [DOI: 10.1007/s11325-016-1435-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/10/2016] [Accepted: 11/07/2016] [Indexed: 11/28/2022]
|
40
|
Microinjection of the melanin-concentrating hormone into the sublaterodorsal tegmental nucleus inhibits REM sleep in the rat. Neurosci Lett 2016; 630:66-69. [DOI: 10.1016/j.neulet.2016.07.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/09/2016] [Accepted: 07/19/2016] [Indexed: 11/20/2022]
|
41
|
Hong KB, Park Y, Suh HJ. Sleep-promoting effects of the GABA/5-HTP mixture in vertebrate models. Behav Brain Res 2016; 310:36-41. [PMID: 27150227 DOI: 10.1016/j.bbr.2016.04.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 04/26/2016] [Accepted: 04/29/2016] [Indexed: 12/11/2022]
Abstract
The aim of this study was to investigate the sleep-promoting effect of combined γ-aminobutyric acid (GABA) and 5-hydroxytryptophan (5-HTP) on sleep quality and quantity in vertebrate models. Pentobarbital-induced sleep test and electroencephalogram (EEG) analysis were applied to investigate sleep latency, duration, total sleeping time and sleep quality of two amino acids and GABA/5-HTP mixture. In addition, real-time PCR and HPLC analysis were applied to analyze the signaling pathway. The GABA/5-HTP mixture significantly regulated the sleep latency, duration (p<0.005), and also increased the sleep quality than single administration of the amino acids (p<0.000). Long-term administration increased the transcript levels of GABAA receptor (1.37-fold, p<0.000) and also increased the GABA content compared with the control group 12h after administration (1.43-fold, p<0.000). Our available evidence suggests that the GABA/5-HTP mixture modulates both GABAergic and serotonergic signaling. Moreover, the sleep architecture can be controlled by the regulation of GABAA receptor and GABA content with 5-HTP.
Collapse
Affiliation(s)
- Ki-Bae Hong
- Department of Public Health Sciences, Korea University, Seoul 136-713, Republic of Korea
| | - Yooheon Park
- Dongguk University Research Institute of Biotechnology, Goyang 10326, Republic of Korea
| | - Hyung Joo Suh
- Department of Public Health Sciences, Korea University, Seoul 136-713, Republic of Korea.
| |
Collapse
|
42
|
Calegare BF, Costa A, Fernandes L, Dias AL, Torterolo P, Almeida VD. Subchronical treatment with Fluoxetine modifies the activity of the MCHergic and hypocretinergic systems. Evidences from peptide CSF concentration and gene expression. Sleep Sci 2016; 9:89-93. [PMID: 27656272 PMCID: PMC5022008 DOI: 10.1016/j.slsci.2016.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/25/2016] [Accepted: 05/31/2016] [Indexed: 01/02/2023] Open
Abstract
In the postero-lateral hypothalamus are located two neuronal systems that utilize the neuropeptides melanin-concentrating hormone (MCH) and hypocretins (also called orexins) as neuromodulators. These systems have reciprocal connections between them, and project throughout the central nervous system. MCH has been involved in the generation of sleep, mainly REM sleep, while hypocretins have a critical role in the generation of wakefulness. MCHergic activity is also involved in the pathophysiology of major depressive disorder (MD). In this regards, intracerebral administration of MCH promotes pro-depressive behaviors (i.e., immobility in the forced swimming test) and REM sleep hypersomnia, which is an important trait of depression. Furthermore, the antagonism of the MCHR-1 receptor has a reliable antidepressant effect, suggesting that MCH is a pro-depressive factor. Hypocretins have been also involved in mood regulation; however, their role in depression is still on debate. Taking these data into account, we explored whether systemic subchronical treatment with Fluoxetine (FLX), a serotonergic antidepressant, modifies the concentration of MCH in the cerebrospinal fluid (CSF), as well as the preproMCH mRNA expression. We also evaluated the hypocretinergic system by quantifying the hypocretin-levels in the CSF and the preprohypocretin mRNA expression. Compared to control, FLX increased the levels of preprohypocretin mRNA without affecting the hypocretin-1 CSF levels. On the contrary, FLX significantly decreased the MCH CSF concentration without affecting the preproMCH gene expression. This result is in agreement with the fact that MCH serum level diminishes during the antidepressant treatment in MD, and supports the hypothesis that an increase in the MCHergic activity could have pro-depressive consequences.
Collapse
Affiliation(s)
- Bruno F. Calegare
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alicia Costa
- Department of Physiology, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Leandro Fernandes
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana L. Dias
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Pablo Torterolo
- Department of Physiology, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Vânia D’ Almeida
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|