1
|
Bell SE, Xie YR, Maciejewski MF, Rubakhin SS, Romanova EV, Bell AM, Sweedler JV. Single-Cell Peptide Profiling to Distinguish Stickleback Ecotypes with Divergent Breeding Behavior. J Proteome Res 2025. [PMID: 39792146 DOI: 10.1021/acs.jproteome.4c00832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Variation in parenting behavior is widespread across the animal kingdom, both within and between species. There are two ecotypes of the three-spined stickleback fish (Gasterosteus aculeatus) that exhibit dramatic differences in their paternal behavior. Males of the common ecotype are highly attentive fathers, tending to young from eggs to fry, while males of the white ecotype desert offspring as eggs. As the pituitary is a key regulator in the hypothalamic-pituitary-adrenal (HPA) axis and the hypothalamic-pituitary-gonadal (HPG) axis between the brain and body, its peptides may influence parenting behaviors. Here, we utilized matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) for high-throughput peptide analysis in single cells of pituitaries from both three-spined stickleback ecotypes. Peptide mass fingerprinting was performed using an in silico generated peptide library to identify detected prohormones. Differential analysis revealed POMC-derived peptides, MCH-derived peptides, and oxytocin as significantly different between the two ecotypes, with higher oxytocin levels in the common ecotype. Interestingly, these subtle chemical differences were not captured by Leiden clustering of the cellular phenotypes. These results call for further investigation of the neurochemical basis for parenting in sticklebacks.
Collapse
Affiliation(s)
- Sara E Bell
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Yuxuan Richard Xie
- Department of Bioengineering and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Meghan F Maciejewski
- Department of Evolution, Ecology, and Behavior, School of Integrative Biology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Stanislav S Rubakhin
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Alison M Bell
- Department of Evolution, Ecology, and Behavior, School of Integrative Biology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana─Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
2
|
Liu X, Yang H, Xu W, Wang X, Tang W, Wang X, Jiao Y, Luan X, Li P, Guo F. Melanin-concentrating hormone attenuates the hedonic feeding induced by orexin-A in the ventral tegmental area of high-fat diet male mice. Front Nutr 2024; 11:1468874. [PMID: 39758319 PMCID: PMC11697430 DOI: 10.3389/fnut.2024.1468874] [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: 07/22/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Objective The ventral tegmental area (VTA), a pivotal hub in the brain's reward circuitry, receives inputs from the lateral hypothalamic area (LHA). However, it remains unclear whether melanin-concentrating hormone (MCH) and orexin-A (OX-A) neurons in the LHA exert individual or cooperative influence on palatable food consumption in the VTA. This study aims to investigate the modulatory role of MCH and OX-A in hedonic feeding within the VTA of high-fat diet (HFD) mice. Methods Male mice were subjected to an 8-week high-fat diet. To visualize the projections from the LHA to VTA, we employed fluorescent gold retrograde tracing combined with immunofluorescence staining. Immunofluorescence staining or enzyme-linked immunosorbent assay was used to detect the activity of the VTA neurons, expression of OX-A or MCH in the LHA, as well as the activity of their receptors (OXR1 and MCHR1) in the VTA following a sucrose preference test. Single-unit extracellular electrical discharge recordings were conducted to assess the effects of OX-A and MCH on VTA neurons in HFD mice. Additionally, chemogenetic inhibition of MCH neurons and immunofluorescence staining were utilized to observe the regulatory roles of MCH in changes of hedonic feeding induced by OX-A in HFD mice. Results Sucrose intake resulted in lower activation of VTA neurons in the HFD mice compared to CON mice, while OX-Aergic and MCHergic neurons project from the LHA to the VTA. Although sucrose intake increased the expression of OX-A and MCH in HFD mice, it led to diminished activation of OXR1-positive and MCHR1-positive VTA neurons compared to CON mice. Extracellular single-unit recording revealed that MCH significantly suppressed the firing rate of OX-A-responsive neurons in the VTA. MCH attenuated the hedonic feeding response induced by OX-A in HFD mice, and administration of MCHR1 antagonist (SNAP94847) significantly potentiated the effect of OX-A. Chemogenetic inhibition of MCH neurons improved the activity of OXR1-expressing neurons, which could be reversed by pretreatment with an OXR1 antagonist (SB334867). Furthermore, chemogenetic inhibition of MCH enhanced hedonic feeding behavior, which was counteracted by SB334867 treatment in HFD mice. Conclusion Melanin-concentrating hormone could attenuate the hedonic feeding behavior induced by orexin-A in the VTA of HFD mice.
Collapse
Affiliation(s)
- Xiaoning Liu
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Pathology, Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Helin Yang
- Department of Spine Surgery, Peking University People’s Hospital, Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Wenguang Xu
- Department of Gastroenterology, Affiliated Qingdao Third People’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Xuezhe Wang
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Wenhui Tang
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiaoxuan Wang
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Yang Jiao
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xinchi Luan
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Pengmeng Li
- Department of Gastroenterology, Affiliated Qingdao Third People’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Feifei Guo
- Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
3
|
Cam Y, Kocum CG, Konrad ER, Schweizer TA, Houska TK, Sardina CA, Suri SK, Will MJ. Incentive motivation for palatable food blocked by intra-accumbens melanin-concentrating hormone (MCH) receptor-1 antagonist in female rats. Pharmacol Biochem Behav 2024; 245:173884. [PMID: 39341509 DOI: 10.1016/j.pbb.2024.173884] [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: 06/19/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Melanin-concentrating hormone (MCH) activity in the nucleus accumbens (Acb) has been shown to influence feeding behavior, yet this has not been characterized in terms of homeostatic vs. hedonic feeding processes. Hedonic feeding, driven by palatability rather than energy deficit, can be modeled through intra-Acb administration of the selective μ-opioid receptor agonist d-Ala2, NMe-Phe4, Glyol5-enkephalin (DAMGO), which preferentially increases consumption and incentive motivation to obtain preferred palatable food. Pharmacological activation of MCH 1 receptors (MCHR1) within Acb has been shown to promote general feeding of chow in males, but not females. However, the effects of MCH on the incentive motivation to obtain preferred palatable food have not been explored. Here, we investigated the role of MCHR1 within the Acb in DAMGO-induced incentive motivation to obtain a sucrose pellet reward. Female Sprague Dawley rats were trained and tested for operant responding under a progressive ratio (PR) breakpoint in response to concurrent intra-Acb administration of DAMGO (0 μg and 0.025 μg/.5 μl/side) immediately following intra-Acb administration of the MCHR1 antagonist (N-(3-{1-[4-(3,4-difluoro-phenoxy)-benzyl]-piperdin-4-yl}-4-methyl-phenyl)-isobutyramide (SNAP-94847; 0 μg, 1.5 μg, and 15 μg/.5 μl/side), in a counterbalanced fashion. As expected, DAMGO significantly increased PR breakpoint and overall active lever presses. SNAP-94847 did not influence PR breakpoint by itself, compared to vehicle; however, both 1.5 and 15 μg doses of SNAP-94847 significantly blocked the increased PR breakpoint produced by intra-Acb DAMGO. The results of the study demonstrate that Acb MCHR1 may play a specific role in the hedonically-driven motivation for palatable food in females.
Collapse
Affiliation(s)
- Yonca Cam
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA.
| | - Courtney G Kocum
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Ella R Konrad
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Tim A Schweizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Tabitha K Houska
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Carlos A Sardina
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Sanya K Suri
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Matthew J Will
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| |
Collapse
|
4
|
Oh JY, Lee H, Jang SY, Kim H, Park G, Serikov A, Jang JH, Kim J, Yang S, Sa M, Lee SE, Han YE, Hwang TY, Jung SJ, Kim HY, Lee SE, Oh SJ, Kim J, Kim J, Kim J, McHugh TJ, Lee CJ, Nam MH, Park HJ. Central Role of Hypothalamic Circuits for Acupuncture's Anti-Parkinsonian Effects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403245. [PMID: 39119926 DOI: 10.1002/advs.202403245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/30/2024] [Indexed: 08/10/2024]
Abstract
Despite clinical data stretching over millennia, the neurobiological basis of the effectiveness of acupuncture in treating diseases of the central nervous system has remained elusive. Here, using an established model of acupuncture treatment in Parkinson's disease (PD) model mice, we show that peripheral acupuncture stimulation activates hypothalamic melanin-concentrating hormone (MCH) neurons via nerve conduction. We further identify two separate neural pathways originating from anatomically and electrophysiologically distinct MCH neuronal subpopulations, projecting to the substantia nigra and hippocampus, respectively. Through chemogenetic manipulation specifically targeting these MCH projections, their respective roles in mediating the acupuncture-induced motor recovery and memory improvements following PD onset are demonstrated, as well as the underlying mechanisms mediating recovery from dopaminergic neurodegeneration, reactive gliosis, and impaired hippocampal synaptic plasticity. Collectively, these MCH neurons constitute not only a circuit-based explanation for the therapeutic effectiveness of traditional acupuncture, but also a potential cellular target for treating both motor and non-motor PD symptoms.
Collapse
Affiliation(s)
- Ju-Young Oh
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyowon Lee
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sun-Young Jang
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Geunhong Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Almas Serikov
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae-Hwan Jang
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Junyeop Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04629, Republic of Korea
| | - Seulkee Yang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Sung Eun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Tae-Yeon Hwang
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sharon Jiyoon Jung
- Technological Convergence Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongyeon Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jongpil Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04629, Republic of Korea
| | - Thomas J McHugh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Laboratory for Circuit and Behavioral Physiology, RIKEN, Wako-shi Saitama, 351-0198, Japan
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hi-Joon Park
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
5
|
Li M. Is melanin-concentrating hormone in the medial preoptic area a signal for the decline of maternal care in late postpartum? Front Neuroendocrinol 2024; 75:101155. [PMID: 39222798 DOI: 10.1016/j.yfrne.2024.101155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
This manuscript proposes that melanin-concentrating hormone (MCH) in the medial preoptic area (MPOA) is an neurochemical signal evolved to trigger the declining process of maternal care. MCH in the MPOA appears only after parturition and is progressively increased with the progression of lactation, while maternal behavior declines progressively. Intra-MPOA injection of MCH decreases active maternal responses. MCH is also highly responsive to infant characteristics and maternal condition. Behavioral changes induced by MCH in late postpartum period are conducive to the decline of infant-directed maternal behavior. The MPOA MCH system may mediate the maternal behavior decline by suppressing the maternal approach motivation and/or increasing maternal withdrawal via its inhibitory action onto the mesolimbic dopamine D1/D2 receptors and its stimulating action on serotonin 5-HT2C receptors in the ventral tegmental area. Research into the MCH maternal effects will enhance our understanding of the neurochemical mechanisms underlying the maternal behavior decline.
Collapse
Affiliation(s)
- Ming Li
- Department of Psychology, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
6
|
Krolick KN, Cao J, Gulla EM, Bhardwaj M, Marshall SJ, Zhou EY, Kiss AJ, Choueiry F, Zhu J, Shi H. Subregion-specific transcriptomic profiling of rat brain reveals sex-distinct gene expression impacted by adolescent stress. Neuroscience 2024; 553:19-39. [PMID: 38977070 PMCID: PMC11444371 DOI: 10.1016/j.neuroscience.2024.07.002] [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: 01/18/2024] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Stress during adolescence clearly impacts brain development and function. Sex differences in adolescent stress-induced or exacerbated emotional and metabolic vulnerabilities could be due to sex-distinct gene expression in hypothalamic, limbic, and prefrontal brain regions. However, adolescent stress-induced whole-genome expression changes in key subregions of these brain regions were unclear. In this study, female and male adolescent Sprague Dawley rats received one-hour restraint stress daily from postnatal day (PD) 32 to PD44. Corticosterone levels, body weights, food intake, body composition, and circulating adiposity and sex hormones were measured. On PD44, brain and blood samples were collected. Using RNA-sequencing, sex-specific differences in stress-induced differentially expressed (DE) genes were identified in subregions of the hypothalamus, limbic system, and prefrontal cortex. Canonical pathways reflected well-known sex-distinct maladies and diseases, substantiating the therapeutic potential of the DE genes found in the current study. Thus, we proposed specific sex distinct, adolescent stress-induced transcriptional changes found in the current study as examples of the molecular bases for sex differences witnessed in stress induced or exacerbated emotional and metabolic disorders. Future behavioral studies and single-cell studies are warranted to test the implications of the DE genes identified in this study in sex-distinct stress-induced susceptibilities.
Collapse
Affiliation(s)
| | - Jingyi Cao
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Evelyn M Gulla
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Meeta Bhardwaj
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | | | - Ethan Y Zhou
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Andor J Kiss
- Center for Bioinformatics & Functional Genomics, Miami University, Oxford, OH 45056, USA.
| | - Fouad Choueiry
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA.
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
7
|
Marinescu AM, Labouesse MA. The nucleus accumbens shell: a neural hub at the interface of homeostatic and hedonic feeding. Front Neurosci 2024; 18:1437210. [PMID: 39139500 PMCID: PMC11319282 DOI: 10.3389/fnins.2024.1437210] [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: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Feeding behavior is a complex physiological process regulated by the interplay between homeostatic and hedonic feeding circuits. Among the neural structures involved, the nucleus accumbens (NAc) has emerged as a pivotal region at the interface of these two circuits. The NAc comprises distinct subregions and in this review, we focus mainly on the NAc shell (NAcSh). Homeostatic feeding circuits, primarily found in the hypothalamus, ensure the organism's balance in energy and nutrient requirements. These circuits monitor peripheral signals, such as insulin, leptin, and ghrelin, and modulate satiety and hunger states. The NAcSh receives input from these homeostatic circuits, integrating information regarding the organism's metabolic needs. Conversely, so-called hedonic feeding circuits involve all other non-hunger and -satiety processes, i.e., the sensory information, associative learning, reward, motivation and pleasure associated with food consumption. The NAcSh is interconnected with hedonics-related structures like the ventral tegmental area and prefrontal cortex and plays a key role in encoding hedonic information related to palatable food seeking or consumption. In sum, the NAcSh acts as a crucial hub in feeding behavior, integrating signals from both homeostatic and hedonic circuits, to facilitate behavioral output via its downstream projections. Moreover, the NAcSh's involvement extends beyond simple integration, as it directly impacts actions related to food consumption. In this review, we first focus on delineating the inputs targeting the NAcSh; we then present NAcSh output projections to downstream structures. Finally we discuss how the NAcSh regulates feeding behavior and can be seen as a neural hub integrating homeostatic and hedonic feeding signals, via a functionally diverse set of projection neuron subpopulations.
Collapse
Affiliation(s)
- Alina-Măriuca Marinescu
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marie A. Labouesse
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
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
|
9
|
Kuebler IRK, Suárez M, Wakabayashi KT. Sex differences and sex-specific regulation of motivated behavior by Melanin-concentrating hormone: a short review. Biol Sex Differ 2024; 15:33. [PMID: 38570844 PMCID: PMC10993549 DOI: 10.1186/s13293-024-00608-0] [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: 11/11/2023] [Accepted: 03/23/2024] [Indexed: 04/05/2024] Open
Abstract
Recent preclinical research exploring how neuropeptide transmitter systems regulate motivated behavior reveal the increasing importance of sex as a critical biological variable. Neuropeptide systems and their central circuits both contribute to sex differences in a range of motivated behaviors and regulate sex-specific behaviors. In this short review, we explore the current research of how sex as a biological variable influences several distinct motivated behaviors that are modulated by the melanin-concentrating hormone (MCH) neuropeptide system. First, we review how MCH regulates feeding behavior within the context of energy homeostasis differently between male and female rodents. Then, we focus on MCH's role in lactation as a sex-specific process within the context of energy homeostasis. Next, we discuss the sex-specific effects of MCH on maternal behavior. Finally, we summarize the role of MCH in drug-motivated behaviors. While these topics are traditionally investigated from different scientific perspectives, in this short review we discuss how these behaviors share commonalities within the larger context of motivated behaviors, and that sex differences discovered in one area of research may impact our understanding in another. Overall, our review highlights the need for further research into how sex differences in energy regulation associated with reproduction and parental care contribute to regulating motivated behaviors.
Collapse
Affiliation(s)
- Isabel R K Kuebler
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, 68588-0308, USA
| | - Mauricio Suárez
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, 68588-0308, USA
| | - Ken T Wakabayashi
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, 68588-0308, USA.
- Rural Drug Addiction Research Center, University of Nebraska-Lincoln, 660 N 12th St., Lincoln, NE, 68588, USA.
| |
Collapse
|
10
|
Kim YK, Jo D, Arjunan A, Ryu Y, Lim YH, Choi SY, Kim HK, Song J. Identification of IGF-1 Effects on White Adipose Tissue and Hippocampus in Alzheimer's Disease Mice via Transcriptomic and Cellular Analysis. Int J Mol Sci 2024; 25:2567. [PMID: 38473814 DOI: 10.3390/ijms25052567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) stands as the most prevalent neurodegenerative disorder, characterized by a multitude of pathological manifestations, prominently marked by the aggregation of amyloid beta. Recent investigations have revealed a compelling association between excessive adiposity and glial activation, further correlating with cognitive impairments. Additionally, alterations in levels of insulin-like growth factor 1 (IGF-1) have been reported in individuals with metabolic conditions accompanied by memory dysfunction. Hence, our research endeavors to comprehensively explore the impact of IGF-1 on the hippocampus and adipose tissue in the context of Alzheimer's disease. To address this, we have conducted an in-depth analysis utilizing APP/PS2 transgenic mice, recognized as a well-established mouse model for Alzheimer's disease. Upon administering IGF-1 injections to the APP/PS2 mice, we observed notable alterations in their behavioral patterns, prompting us to undertake a comprehensive transcriptomic analysis of both the hippocampal and adipose tissues. Our data unveiled significant modifications in the functional profiles of these tissues. Specifically, in the hippocampus, we identified changes associated with synaptic activity and neuroinflammation. Concurrently, the adipose tissue displayed shifts in processes related to fat browning and cell death signaling. In addition to these findings, our analysis enabled the identification of a collection of long non-coding RNAs and circular RNAs that exhibited significant changes in expression subsequent to the administration of IGF-1 injections. Furthermore, we endeavored to predict the potential roles of these identified RNA molecules within the context of our study. In summary, our study offers valuable transcriptome data for hippocampal and adipose tissues within an Alzheimer's disease model and posits a significant role for IGF-1 within both the hippocampus and adipose tissue.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Danbi Jo
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Yeongseo Ryu
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Yeong-Hwan Lim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Seo Yoon Choi
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Hee Kyung Kim
- Department of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Juhyun Song
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| |
Collapse
|
11
|
Everett T, Ten Eyck TW, Wu CH, Shelowitz AL, Stansbury SM, Firek A, Setlow B, McIntyre JC. Cilia loss on distinct neuron populations differentially alters cocaine-induced locomotion and reward. J Psychopharmacol 2024; 38:200-212. [PMID: 38151883 PMCID: PMC11078551 DOI: 10.1177/02698811231219058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuronal primary cilia are being recognized for their role in mediating signaling associated with a variety of neurobehaviors, including responses to drugs of abuse. They function as signaling hubs, enriched with a diverse array of G-protein coupled receptors (GPCRs), including several associated with motivation and drug-related behaviors. However, our understanding of how cilia regulate neuronal function and behavior is still limited. AIMS The objective of the current study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to cocaine. METHODS To test the consequences of cilia loss on cocaine-induced locomotion and reward-related behavior, we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. RESULTS Cilia ablation on either population of neurons failed to significantly alter acute locomotor responses to cocaine at a range of doses. With repeated administration, mice lacking cilia on GAD2-GABAergic neurons showed no difference in locomotor sensitization to cocaine compared to wild-type (WT) littermates, whereas mice lacking cilia on dopaminergic neurons exhibited reduced locomotor sensitization to cocaine at 10 and 30 mg/kg. Mice lacking cilia on GAD2-GABAergic neurons showed no difference in cocaine conditioned place preference (CPP), whereas mice lacking cilia on dopaminergic neurons exhibited reduced CPP compared to WT littermates. CONCLUSIONS Combined with previous findings using amphetamine, our results show that behavioral effects of cilia ablation are cell- and drug type-specific, and that neuronal cilia contribute to modulation of both the locomotor-inducing and rewarding properties of cocaine.
Collapse
Affiliation(s)
- Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Tyler W. Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Chang-Hung Wu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | | | - Sofia M. Stansbury
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Alexandra Firek
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| |
Collapse
|
12
|
Miller PA, Williams-Ikhenoba JG, Sankhe AS, Hoffe BH, Chee MJ. Neuroanatomical, electrophysiological, and morphological characterization of melanin-concentrating hormone cells coexpressing cocaine- and amphetamine-regulated transcript. J Comp Neurol 2024; 532:e25588. [PMID: 38335050 DOI: 10.1002/cne.25588] [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: 05/01/2023] [Revised: 12/18/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
Melanin-concentrating hormone (MCH) cells in the hypothalamus regulate fundamental physiological functions like energy balance, sleep, and reproduction. This diversity may be ascribed to the neurochemical heterogeneity among MCH cells. One prominent subpopulation of MCH cells coexpresses cocaine- and amphetamine-regulated transcript (CART), and as MCH and CART can have opposing actions, MCH/CART+ and MCH/CART- cells may differentially modulate behavioral outcomes. However, it is not known if there are differences in the cellular properties underlying their functional differences; thus, we compared the neuroanatomical, electrophysiological, and morphological properties of MCH cells in male and female Mch-cre;L10-Egfp reporter mice. Half of MCH cells expressed CART and were most prominent in the medial hypothalamus. Whole-cell patch-clamp recordings revealed differences in their passive and active membrane properties in a sex-dependent manner. Female MCH/CART+ cells had lower input resistances, but male cells largely differed in their firing properties. All MCH cells increased firing when stimulated, but their firing frequency decreases with sustained stimulation. MCH/CART+ cells showed stronger spike rate adaptation than MCH/CART- cells. The kinetics of excitatory events at MCH cells also differed by cell type, as the rising rate of excitatory events was slower at MCH/CART+ cells. By reconstructing the dendritic arborization of our recorded cells, we found no sex differences, but male MCH/CART+ cells had less dendritic length and fewer branch points. Overall, distinctions in topographical division and cellular properties between MCH cells add to their heterogeneity and help elucidate their response to stimuli or effect on modulating their respective neural networks.
Collapse
Affiliation(s)
| | | | - Aditi S Sankhe
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Brendan H Hoffe
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
13
|
Kuebler IRK, Liu Y, Bueno Álvarez BS, Huber NM, Jolton JA, Dasari R, Wakabayashi KT. Melanin-concentrating hormone receptor antagonism differentially attenuates nicotine experience-dependent locomotor behavior in female and male rats. Pharmacol Biochem Behav 2023; 232:173649. [PMID: 37793486 PMCID: PMC10985048 DOI: 10.1016/j.pbb.2023.173649] [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: 07/18/2023] [Revised: 09/14/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
Nicotine is a significant public health concern because it is the primary pharmacological agent in tobacco use disorder. One neural system that has been implicated in the symptoms of several substance use disorders is the melanin-concentrating hormone (MCH) system. MCH regulates various motivated behaviors depending on sex, yet little is known of how this interaction affects experience with drugs of abuse, particularly nicotine. The goal of this study was to determine the effect of MCH receptor antagonism on experience-dependent nicotine-induced locomotion after chronic exposure, particularly on the expression of locomotor sensitization. Adult female and male Wistar rats were given saline then cumulative doses of nicotine (0.1, 0.32, 0.56, and 1.0 mg/kg) intraperitoneally to determine the acute effects of nicotine (day 1). Next, rats were treated with 1.0 mg/kg nicotine for 6 days, given an identical series of cumulative doses (day 8), and then kept in a drug-free state for 6 days. On day 15, rats were pretreated with vehicle or the MCH receptor antagonist GW803430 (10 or 30 mg/kg) before another series of cumulative doses to assess response to chronic nicotine. After vehicle, male rats increased nicotine locomotor activation from day 1 to day 15, and both sexes showed a sensitized response when normalized to saline. The lower dose of GW803430 decreased locomotion compared to vehicle in females, while the higher dose decreased locomotion in males. Both sexes showed nicotine dose-dependent effects of GW803430, strongest at lower doses of nicotine. Controlling for sex-based locomotor differences revealed that females are more sensitive to GW803430. The high dose of GW803430 also decreased saline locomotion in males. Together, the results of our study suggest that MCH is involved in the expression of nicotine locomotor sensitization, and that MCH regulates these nicotine behavioral symptoms differently across sex.
Collapse
Affiliation(s)
- Isabel R K Kuebler
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America
| | - Youxi Liu
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America
| | - Bárbara S Bueno Álvarez
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America
| | - Noah M Huber
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America
| | - Joshua A Jolton
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America
| | - Raaga Dasari
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America
| | - Ken T Wakabayashi
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, 1220 T St., Lincoln, NE 68588, United States of America; Rural Drug Addiction Research Center, University of Nebraska-Lincoln, 660 N 12th St., Lincoln, NE 68588, United States of America.
| |
Collapse
|
14
|
Ma L, Liu H, Xu Z, Yang M, Zhang Y. Application of the wholebrain calculation interactive framework to map whole-brain neural connectivity networks. J Chem Neuroanat 2023; 132:102304. [PMID: 37331669 DOI: 10.1016/j.jchemneu.2023.102304] [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: 04/20/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The aim of this work was to develop a simple and feasible method of mapping the neural network topology of the mouse brain. Wild-type C57BL/6 J mice (n = 10) aged 8-10 weeks were injected with the cholera toxin subunit B (CTB) tracer in the anterior (NAcCA) and posterior (NAcCP) parts of the nucleus accumbens (NAc) core and in the medial (NAcSM) and lateral (NAcSL) parts of the NAc shell. The labeled neurons were reconstructed using the WholeBrain Calculation Interactive Framework. The NAcCA receives neuronal projections from the olfactory areas (OLF) and isocortex; the thalamus and isocortex project more fibers to the NAcSL, and the hypothalamus send more fiber projections to the NAcSM. Cell resolution can be automatically annotated, analyzed, and visualized using the WholeBrain Calculation Interactive Framework, making large-scale mapping of mouse brains at cellular and subcellular resolutions easier and more accurate.
Collapse
Affiliation(s)
- Liping Ma
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - He Liu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Ziyi Xu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Mengli Yang
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Yinghua Zhang
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
15
|
Calafate S, Özturan G, Thrupp N, Vanderlinden J, Santa-Marinha L, Morais-Ribeiro R, Ruggiero A, Bozic I, Rusterholz T, Lorente-Echeverría B, Dias M, Chen WT, Fiers M, Lu A, Vlaeminck I, Creemers E, Craessaerts K, Vandenbempt J, van Boekholdt L, Poovathingal S, Davie K, Thal DR, Wierda K, Oliveira TG, Slutsky I, Adamantidis A, De Strooper B, de Wit J. Early alterations in the MCH system link aberrant neuronal activity and sleep disturbances in a mouse model of Alzheimer's disease. Nat Neurosci 2023:10.1038/s41593-023-01325-4. [PMID: 37188873 DOI: 10.1038/s41593-023-01325-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
Early Alzheimer's disease (AD) is associated with hippocampal hyperactivity and decreased sleep quality. Here we show that homeostatic mechanisms transiently counteract the increased excitatory drive to CA1 neurons in AppNL-G-F mice, but that this mechanism fails in older mice. Spatial transcriptomics analysis identifies Pmch as part of the adaptive response in AppNL-G-F mice. Pmch encodes melanin-concentrating hormone (MCH), which is produced in sleep-active lateral hypothalamic neurons that project to CA1 and modulate memory. We show that MCH downregulates synaptic transmission, modulates firing rate homeostasis in hippocampal neurons and reverses the increased excitatory drive to CA1 neurons in AppNL-G-F mice. AppNL-G-F mice spend less time in rapid eye movement (REM) sleep. AppNL-G-F mice and individuals with AD show progressive changes in morphology of CA1-projecting MCH axons. Our findings identify the MCH system as vulnerable in early AD and suggest that impaired MCH-system function contributes to aberrant excitatory drive and sleep defects, which can compromise hippocampus-dependent functions.
Collapse
Affiliation(s)
- Sara Calafate
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Gökhan Özturan
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Nicola Thrupp
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luísa Santa-Marinha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Bozic
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Thomas Rusterholz
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Blanca Lorente-Echeverría
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Marcelo Dias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Wei-Ting Chen
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ashley Lu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ine Vlaeminck
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Eline Creemers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Joris Vandenbempt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luuk van Boekholdt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
- KU Leuven, Department of Otorhinolaryngology, Leuven, Belgium
| | - Suresh Poovathingal
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Kristofer Davie
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory of Neuropathology, and Leuven Brain Institute, KU-Leuven, O&N IV, Leuven, Belgium
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Keimpe Wierda
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Antoine Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- UK Dementia Research Institute (UK DRI@UCL) at University College London, London, UK.
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
| |
Collapse
|
16
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
17
|
Ruiz-Viroga V, de Ceglia M, Morelli L, Castaño EM, Calvo EB, Suárez J, Rodríguez de Fonseca F, Galeano P, Lagos P. Acute intrahippocampal administration of melanin-concentrating hormone impairs memory consolidation and decreases the expression of MCHR-1 and TrkB receptors. Prog Neuropsychopharmacol Biol Psychiatry 2023; 123:110703. [PMID: 36565982 DOI: 10.1016/j.pnpbp.2022.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Interest in the role of melanin-concentrating hormone (MCH) in memory processes has increased in recent years, with some studies reporting memory-enhancing effects, while others report deleterious effects. Due to these discrepancies, this study seeks to provide new evidence about the role of MCH in memory consolidation and its relation with BDNF/TrkB system. To this end, in the first experiment, increased doses of MCH were acutely administered in both hippocampi to groups of male rats (25, 50, 200, and 500 ng). Microinjections were carried out immediately after finishing the sample trial of two hippocampal-dependent behavioral tasks: the Novel Object Recognition Test (NORT) and the modified Elevated Plus Maze (mEPM) test. Results indicated that a dose of 200 ng of MCH or higher impaired memory consolidation in both tasks. A second experiment was performed in which a dose of 200 ng of MCH was administered alone or co-administered with the MCHR-1 antagonist ATC-0175 at the end of the sample trial in the NORT. Results showed that MCH impaired memory consolidation, while the co-administration with ATC-0175 reverted this detrimental effect. Moreover, MCH induced a significant decrease in hippocampal MCHR-1 and TrkB expression with no modification in the expression of BDNF and NMDA receptor subunits NR1, NR2A, and NR2B. These results suggest that MCH in vivo elicits pro-amnesic effects in the rat hippocampus by decreasing the availability of its receptor and TrkB receptors, thus linking both endogenous systems to memory processes.
Collapse
Affiliation(s)
- Vicente Ruiz-Viroga
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay
| | - Marialuisa de Ceglia
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo M Castaño
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo Blanco Calvo
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Campus de Teatinos S/N, Málaga 29071, Spain.
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Málaga 29071, Spain.
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Patricia Lagos
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay.
| |
Collapse
|
18
|
Vas S, Papp RS, Könczöl K, Bogáthy E, Papp N, Ádori C, Durst M, Sípos K, Ocskay K, Farkas I, Bálint F, Ferenci S, Török B, Kovács A, Szabó E, Zelena D, Kovács KJ, Földes A, Kató E, Köles L, Bagdy G, Palkovits M, Tóth ZE. Prolactin-Releasing Peptide Contributes to Stress-Related Mood Disorders and Inhibits Sleep/Mood Regulatory Melanin-Concentrating Hormone Neurons in Rats. J Neurosci 2023; 43:846-862. [PMID: 36564184 PMCID: PMC9899089 DOI: 10.1523/jneurosci.2139-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 08/31/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Stress disorders impair sleep and quality of life; however, their pathomechanisms are unknown. Prolactin-releasing peptide (PrRP) is a stress mediator; we therefore hypothesized that PrRP may be involved in the development of stress disorders. PrRP is produced by the medullary A1/A2 noradrenaline (NA) cells, which transmit stress signals to forebrain centers, and by non-NA cells in the hypothalamic dorsomedial nucleus. We found in male rats that both PrRP and PrRP-NA cells innervate melanin-concentrating hormone (MCH) producing neurons in the dorsolateral hypothalamus (DLH). These cells serve as a key hub for regulating sleep and affective states. Ex vivo, PrRP hyperpolarized MCH neurons and further increased the hyperpolarization caused by NA. Following sleep deprivation, intracerebroventricular PrRP injection reduced the number of REM sleep-active MCH cells. PrRP expression in the dorsomedial nucleus was upregulated by sleep deprivation, while downregulated by REM sleep rebound. Both in learned helplessness paradigm and after peripheral inflammation, impaired coping with sustained stress was associated with (1) overactivation of PrRP cells, (2) PrRP protein and receptor depletion in the DLH, and (3) dysregulation of MCH expression. Exposure to stress in the PrRP-insensitive period led to increased passive coping with stress. Normal PrRP signaling, therefore, seems to protect animals against stress-related disorders. PrRP signaling in the DLH is an important component of the PrRP's action, which may be mediated by MCH neurons. Moreover, PrRP receptors were downregulated in the DLH of human suicidal victims. As stress-related mental disorders are the leading cause of suicide, our findings may have particular translational relevance.SIGNIFICANCE STATEMENT Treatment resistance to monoaminergic antidepressants is a major problem. Neuropeptides that modulate the central monoaminergic signaling are promising targets for developing alternative therapeutic strategies. We found that stress-responsive prolactin-releasing peptide (PrRP) cells innervated melanin-concentrating hormone (MCH) neurons that are crucial in the regulation of sleep and mood. PrRP inhibited MCH cell activity and enhanced the inhibitory effect evoked by noradrenaline, a classic monoamine, on MCH neurons. We observed that impaired PrRP signaling led to failure in coping with chronic/repeated stress and was associated with altered MCH expression. We found alterations of the PrRP system also in suicidal human subjects. PrRP dysfunction may underlie stress disorders, and fine-tuning MCH activity by PrRP may be an important part of the mechanism.
Collapse
Affiliation(s)
- Szilvia Vas
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Semmelweis University, Budapest, 1089, Hungary
| | - Rege S Papp
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, 1094, Hungary
| | - Katalin Könczöl
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Emese Bogáthy
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
| | - Noémi Papp
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
| | - Csaba Ádori
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Máté Durst
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Klaudia Sípos
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Klementina Ocskay
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Flóra Bálint
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Szilamér Ferenci
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Bibiána Török
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Anita Kovács
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Evelin Szabó
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Dóra Zelena
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Krisztina J Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, 1083, Hungary
| | - Anna Földes
- Department of Oral Biology, Semmelweis University, Budapest, 1089, Hungary
| | - Erzsébet Kató
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - László Köles
- Department of Oral Biology, Semmelweis University, Budapest, 1089, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - György Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, 1089, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Semmelweis University, Budapest, 1089, Hungary
- NAP2-SE New Antidepressant Target Research Group, Budapest, 1085, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, 1094, Hungary
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Zsuzsanna E Tóth
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
19
|
Kobayashi Y, Saito Y. Evaluation of ciliary-GPCR dynamics using a validated organotypic brain slice culture method. Methods Cell Biol 2023; 175:69-83. [PMID: 36967146 DOI: 10.1016/bs.mcb.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The primary cilium is a structural organelle present in most mammalian cells. Primary cilia are enriched with a unique protein repertoire distinct from that of the cytosol and the plasma membrane. Such a highly organized microenvironment creates effective machinery for translating extracellular cues into intracellular signals. G protein-coupled receptors (GPCRs) are key receptors in sensing environmental stimuli transmitted via a second messenger into a cellular response. Recent data has demonstrated that a limited number of non-olfactory GPCRs, including melanin-concentrating hormone receptor 1 (MCHR1), are preferentially localized to ciliary membranes of several mammalian cell types, including neuronal cells. Evidence was accumulated to support the functional importance of ciliary-GPCR signaling accompanying ciliary structural changes using cilia-specific cell and molecular biology techniques. Thus, cilia are now considered to function as a unique sensory platform for the integration of GPCR signaling and various cytoplasmic domains. Dissociated neurons expressing ciliary-GPCRs can be a useful tool for examining ciliary dynamics. However, losing preexisting neuronal connectivity may alter neuronal ciliary morphology, such as abnormal elongation. Brain slices prepared under ex vitro conditions are a powerful approach that maintains the cytoarchitecture, enabling researchers to have accurate control over experimental conditions and to study individual cells from subregions of the brain. Here, we present a detailed description of our novel modified method for organotypic culture of rat brain slice and a validated immunostaining protocol to characterize ciliary-GPCR dynamics in coupling with neuropeptides or aminergic activation.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Yumiko Saito
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
20
|
Wong SG, Vorakunthada Y, Lee-Iannotti J, Johnson KG. Sleep-related motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:383-397. [PMID: 37562879 DOI: 10.1016/b978-0-323-98818-6.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Sleep-related motor disorders include non-rapid-eye movement (NREM) sleep parasomnias, rapid-eye movement (REM), sleep parasomnias including REM sleep behavior disorder (RBD), isolated motor phenomena in sleep, and periodic limb movement disorder. Restless legs syndrome (RLS) occurs while awake but is closely related to sleep and has a circadian pattern. The pontine sublaterodorsal tegmental nucleus has an important role in aligning motor control with sleep states, and dysfunction in this region can explain motor activities including cataplexy and loss of REM atonia seen in REM sleep behavior disorder. This chapter begins with a review of motor control in sleep. The rest of the chapter summarizes the clinical presentation, epidemiology, differential and treatment of NREM, REM, and isolated sleep-related motor disorders as well as restless legs syndrome.
Collapse
Affiliation(s)
- Stephanie G Wong
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Yuttiwat Vorakunthada
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Joyce Lee-Iannotti
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Karin G Johnson
- Department of Neurology, University of Massachusetts Chan School of Medicine-Baystate, Springfield, MA, United States; Institute for Healthcare Delivery and Population Science, University of Massachusetts Chan School of Medicine-Baystate, Springfield, MA, United States.
| |
Collapse
|
21
|
Potter LE, Burgess CR. The melanin-concentrating hormone system as a target for the treatment of sleep disorders. Front Neurosci 2022; 16:952275. [PMID: 36177357 PMCID: PMC9513178 DOI: 10.3389/fnins.2022.952275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Given the widespread prevalence of sleep disorders and their impacts on health, it is critical that researchers continue to identify and evaluate novel avenues of treatment. Recently the melanin-concentrating hormone (MCH) system has attracted commercial and scientific interest as a potential target of pharmacotherapy for sleep disorders. This interest emerges from basic scientific research demonstrating a role for MCH in regulating sleep, and particularly REM sleep. In addition to this role in sleep regulation, the MCH system and the MCH receptor 1 (MCHR1) have been implicated in a wide variety of other physiological functions and behaviors, including feeding/metabolism, reward, anxiety, depression, and learning. The basic research literature on sleep and the MCH system, and the history of MCH drug development, provide cause for both skepticism and cautious optimism about the prospects of MCH-targeting drugs in sleep disorders. Extensive efforts have focused on developing MCHR1 antagonists for use in obesity, however, few of these drugs have advanced to clinical trials, and none have gained regulatory approval. Additional basic research will be needed to fully characterize the MCH system’s role in sleep regulation, for example, to fully differentiate between MCH-neuron and peptide/receptor-mediated functions. Additionally, a number of issues relating to drug design will continue to pose a practical challenge for novel pharmacotherapies targeting the MCH system.
Collapse
Affiliation(s)
- Liam E. Potter
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Liam E. Potter,
| | - Christian R. Burgess
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- Christian R. Burgess,
| |
Collapse
|
22
|
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: 0.3] [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
|
23
|
Croizier S, Bouret SG. Molecular Control of the Development of Hypothalamic Neurons Involved in Metabolic Regulation. J Chem Neuroanat 2022; 123:102117. [DOI: 10.1016/j.jchemneu.2022.102117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/03/2022] [Accepted: 06/03/2022] [Indexed: 10/18/2022]
|
24
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
25
|
Gaspari S, Quenneville S, Rodriguez Sanchez‐Archidona A, Thorens B, Croizier S. Structural and molecular characterization of paraventricular thalamic glucokinase-expressing neuronal circuits in the mouse. J Comp Neurol 2022; 530:1773-1949. [PMID: 35303367 PMCID: PMC9542162 DOI: 10.1002/cne.25312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/11/2022]
Abstract
The thalamic paraventricular nucleus (PVT) is a structure highly interconnected with several nuclei ranging from forebrain to hypothalamus and brainstem. Numerous rodent studies have examined afferent and efferent connections of the PVT and their contribution to behavior, revealing its important role in the integration of arousal cues. However, the majority of these studies used a region‐oriented approach, without considering the neuronal subtype diversity of the nucleus. In the present study, we provide the anatomical and transcriptomic characterization of a subpopulation of PVT neurons molecularly defined by the expression of glucokinase (Gck). Combining a genetically modified mouse model with viral tracing approaches, we mapped both the anterograde and the retrograde projections of Gck‐positive neurons of the anterior PVT (GckaPVT). Our results demonstrated that GckaPVT neurons innervate several nuclei throughout the brain axis. The strongest connections are with forebrain areas associated with reward and stress and with hypothalamic structures involved in energy balance and feeding regulation. Furthermore, transcriptomic analysis of the Gck‐expressing neurons revealed that they are enriched in receptors for hypothalamic‐derived neuropeptides, adhesion molecules, and obesity and diabetes susceptibility transcription factors. Using retrograde labeling combined with immunohistochemistry and in situ hybridization, we identify that GckaPVT neurons receive direct inputs from well‐defined hypothalamic populations, including arginine‐vasopressin‐, melanin‐concentrating hormone‐, orexin‐, and proopiomelanocortin‐expressing neurons. This detailed anatomical and transcriptomic characterization of GckaPVT neurons provides a basis for functional studies of the integration of homeostatic and hedonic aspects of energy homeostasis, and for deciphering the potential role of these neurons in obesity and diabetes development.
Collapse
Affiliation(s)
- Sevasti Gaspari
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Simon Quenneville
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | | | - Bernard Thorens
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Sophie Croizier
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
26
|
Bell SM, Evans JM, Evans KM, Tsai KL, Noorai RE, Famula TR, Holle DM, Clark LA. Congenital idiopathic megaesophagus in the German shepherd dog is a sex-differentiated trait and is associated with an intronic variable number tandem repeat in Melanin-Concentrating Hormone Receptor 2. PLoS Genet 2022; 18:e1010044. [PMID: 35271580 PMCID: PMC8912139 DOI: 10.1371/journal.pgen.1010044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/20/2022] [Indexed: 11/19/2022] Open
Abstract
Congenital idiopathic megaesophagus (CIM) is a gastrointestinal (GI) motility disorder of dogs in which reduced peristaltic activity and dilation of the esophagus prevent the normal transport of food into the stomach. Affected puppies regurgitate meals and water, fail to thrive, and experience complications such as aspiration pneumonia that may necessitate euthanasia. The German shepherd dog (GSD) has the highest disease incidence, indicative of a genetic predisposition. Here, we discover that male GSDs are twice as likely to be affected as females and show that the sex bias is independent of body size. We propose that female endogenous factors (e.g., estrogen) are protective via their role in promoting relaxation of the sphincter between the esophagus and stomach, facilitating food passage. A genome-wide association study for CIM revealed an association on canine chromosome 12 (P-val = 3.12x10-13), with the lead SNPs located upstream or within Melanin-Concentrating Hormone Receptor 2 (MCHR2), a compelling positional candidate gene having a role in appetite, weight, and GI motility. Within the first intron of MCHR2, we identified a 33 bp variable number tandem repeat (VNTR) containing a consensus binding sequence for the T-box family of transcription factors. Across dogs and wolves, the major allele includes two copies of the repeat, whereas the predominant alleles in GSDs have one or three copies. The single-copy allele is strongly associated with CIM (P-val = 1.32x10-17), with homozygosity for this allele posing the most significant risk. Our findings suggest that the number of T-box protein binding motifs may correlate with MCHR2 expression and that an imbalance of melanin-concentrating hormone plays a role in CIM. We describe herein the first genetic factors identified in CIM: sex and a major locus on chromosome 12, which together predict disease state in the GSD with greater than 75% accuracy. German shepherd dogs (GSDs) are predisposed to an inherited motility disorder of the esophagus, termed congenital idiopathic megaesophagus (CIM), in which swallowing is ineffective and the esophagus is enlarged. Affected puppies are unable to properly pass food into their stomachs and consequently regurgitate their meals and show a failure to thrive, often leading to euthanasia. Here, we discovered that male GSDs are affected at a ratio of almost 2-to-1 over females, suggesting a protective biological advantage in females. In humans, estrogen is thought to play a role in the male predominance of esophageal disorders like reflux esophagitis and esophageal cancer. In a genome-wide scan, we identified an association with CIM on chromosome 12 and, within this region, a repetitive sequence in MCHR2. This gene encodes a receptor for melanin-concentrating hormone, a signaling molecule that is linked to appetite, weight, and gut motility. Together, sex and the MCHR2 repeat sequence accurately predict affection status in over 75% of dogs, and a genetic test is now available to facilitate breeding decisions aimed at reducing disease incidence.
Collapse
Affiliation(s)
- Sarah M. Bell
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Jacquelyn M. Evans
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Katy M. Evans
- The Seeing Eye Inc., Morristown, New Jersey, United States of America
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Kate L. Tsai
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Rooksana E. Noorai
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
- Clemson University Genomics and Bioinformatics Facility, Clemson University, Clemson, South Carolina, United States of America
| | - Thomas R. Famula
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Dolores M. Holle
- The Seeing Eye Inc., Morristown, New Jersey, United States of America
| | - Leigh Anne Clark
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
27
|
Characterization of Hypothalamic MCH Neuron Development in a 3D Differentiation System of Mouse Embryonic Stem Cells. eNeuro 2022; 9:ENEURO.0442-21.2022. [PMID: 35437265 PMCID: PMC9047030 DOI: 10.1523/eneuro.0442-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 01/20/2023] Open
Abstract
Hypothalamic melanin-concentrating hormone (MCH) neurons are important regulators of multiple physiological processes, such as sleep, feeding, and memory. Despite the increasing interest in their neuronal functions, the molecular mechanism underlying MCH neuron development remains poorly understood. We report that a three-dimensional culture of mouse embryonic stem cells (mESCs) can generate hypothalamic-like tissues containing MCH-positive neurons, which reproduce morphologic maturation, neuronal connectivity, and neuropeptide/neurotransmitter phenotype of native MCH neurons. Using this in vitro system, we demonstrate that Hedgehog (Hh) signaling serves to produce major neurochemical subtypes of MCH neurons characterized by the presence or absence of cocaine- and amphetamine-regulated transcript (CART). Without exogenous Hh signals, mESCs initially differentiated into dorsal hypothalamic/prethalamic progenitors and finally into MCH+CART+ neurons through a specific intermediate progenitor state. Conversely, activation of the Hh pathway specified ventral hypothalamic progenitors that generate both MCH+CART− and MCH+CART+ neurons. These results suggest that in vivo MCH neurons may originate from multiple cell lineages that arise through early dorsoventral patterning of the hypothalamus. Additionally, we found that Hh signaling supports the differentiation of mESCs into orexin/hypocretin neurons, a well-defined cell group intermingled with MCH neurons in the lateral hypothalamic area (LHA). The present study highlights and improves the utility of mESC culture in the analysis of the developmental programs of specific hypothalamic cell types.
Collapse
|
28
|
Miller CL. The Epigenetics of Psychosis: A Structured Review with Representative Loci. Biomedicines 2022; 10:561. [PMID: 35327363 PMCID: PMC8945330 DOI: 10.3390/biomedicines10030561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 02/04/2023] Open
Abstract
The evidence for an environmental component in chronic psychotic disorders is strong and research on the epigenetic manifestations of these environmental impacts has commenced in earnest. In reviewing this research, the focus is on three genes as models for differential methylation, MCHR1, AKT1 and TDO2, each of which have been investigated for genetic association with psychotic disorders. Environmental factors associated with psychotic disorders, and which interact with these model genes, are explored in depth. The location of transcription factor motifs relative to key methylation sites is evaluated for predicted gene expression results, and for other sites, evidence is presented for methylation directing alternative splicing. Experimental results from key studies show differential methylation: for MCHR1, in psychosis cases versus controls; for AKT1, as a pre-existing methylation pattern influencing brain activation following acute administration of a psychosis-eliciting environmental stimulus; and for TDO2, in a pattern associated with a developmental factor of risk for psychosis, in all cases the predicted expression impact being highly dependent on location. Methylation induced by smoking, a confounding variable, exhibits an intriguing pattern for all three genes. Finally, how differential methylation meshes with Darwinian principles is examined, in particular as it relates to the "flexible stem" theory of evolution.
Collapse
|
29
|
Millard SJ, Bearden CE, Karlsgodt KH, Sharpe MJ. The prediction-error hypothesis of schizophrenia: new data point to circuit-specific changes in dopamine activity. Neuropsychopharmacology 2022; 47:628-640. [PMID: 34588607 PMCID: PMC8782867 DOI: 10.1038/s41386-021-01188-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a severe psychiatric disorder affecting 21 million people worldwide. People with schizophrenia suffer from symptoms including psychosis and delusions, apathy, anhedonia, and cognitive deficits. Strikingly, schizophrenia is characterised by a learning paradox involving difficulties learning from rewarding events, whilst simultaneously 'overlearning' about irrelevant or neutral information. While dysfunction in dopaminergic signalling has long been linked to the pathophysiology of schizophrenia, a cohesive framework that accounts for this learning paradox remains elusive. Recently, there has been an explosion of new research investigating how dopamine contributes to reinforcement learning, which illustrates that midbrain dopamine contributes in complex ways to reinforcement learning, not previously envisioned. This new data brings new possibilities for how dopamine signalling contributes to the symptomatology of schizophrenia. Building on recent work, we present a new neural framework for how we might envision specific dopamine circuits contributing to this learning paradox in schizophrenia in the context of models of reinforcement learning. Further, we discuss avenues of preclinical research with the use of cutting-edge neuroscience techniques where aspects of this model may be tested. Ultimately, it is hoped that this review will spur to action more research utilising specific reinforcement learning paradigms in preclinical models of schizophrenia, to reconcile seemingly disparate symptomatology and develop more efficient therapeutics.
Collapse
Affiliation(s)
- Samuel J. Millard
- grid.19006.3e0000 0000 9632 6718Department of Psychology, University of California, Los Angeles, CA 90095 USA
| | - Carrie E. Bearden
- grid.19006.3e0000 0000 9632 6718Department of Psychology, University of California, Los Angeles, CA 90095 USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA 90095 USA
| | - Katherine H. Karlsgodt
- grid.19006.3e0000 0000 9632 6718Department of Psychology, University of California, Los Angeles, CA 90095 USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA 90095 USA
| | - Melissa J. Sharpe
- grid.19006.3e0000 0000 9632 6718Department of Psychology, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
30
|
Huszár J, Bozó É, Beke G, Szalai KK, Kardos P, Boros A, Greiner I, Éles J. hERG optimization of MCHR1 antagonist benzofuro-pyridine and pyrazino-indole derivatives. ChemMedChem 2022; 17:e202100707. [PMID: 35041296 DOI: 10.1002/cmdc.202100707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/21/2021] [Indexed: 11/09/2022]
Abstract
Obesity is a global epidemic associated with multiple severe diseases. Several pharmacotherapies have been investigated including the antagonists of melanin concentrating hormone receptor 1 (MCHR1). The design, synthesis, and biological studies of novel MCHR1 antagonists based on benzofuro-pyridine and pyrazino-indole scaffold was performed. We confirmed that fine-tuning lipophilicity and basic pKa by modifying the benzyl-group and introducing different substituents on the aliphatic nitrogen sidechain decreases both hERG inhibition and metabolic clearance. We have succeeded to develop excellent in vitro parameters in case of compounds 17 and 23g , that can be considered as valuable tools for further pharmacological investigation.
Collapse
Affiliation(s)
- József Huszár
- Gedeon Richter PLc., Department of chemistry, Gyömrői út 19-21, 1103, Budapest, HUNGARY
| | - Éva Bozó
- Gedeon Richter Nyrt, Department of Chemistry, HUNGARY
| | - Gyula Beke
- Gedeon Richter Nyrt, Department of Chemistry, HUNGARY
| | | | - Péter Kardos
- Gedeon Richter Nyrt, Pharmacological and Drug Safety Research, HUNGARY
| | - András Boros
- Gedeon Richter Nyrt, Pharmacological and Drug Safety Research, HUNGARY
| | | | - János Éles
- Gedeon Richter Nyrt, Department of Chemistry, HUNGARY
| |
Collapse
|
31
|
Lanzillo M, Gervais M, Croizier S. Ontogeny of the Projections From the Dorsomedial Division of the Anterior Bed Nucleus of the Stria Terminalis to Hypothalamic Nuclei. Front Neurosci 2021; 15:748186. [PMID: 34916896 PMCID: PMC8669758 DOI: 10.3389/fnins.2021.748186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/27/2021] [Indexed: 12/01/2022] Open
Abstract
The bed nucleus of the stria terminalis (BNST) is a telencephalic structure well-connected to hypothalamic regions known to control goal-oriented behaviors such as feeding. In particular, we showed that the dorsomedial division of the anterior BNST innervate neurons of the paraventricular (PVH), dorsomedial (DMH), and arcuate (ARH) hypothalamic nuclei as well as the lateral hypothalamic area (LHA). While the anatomy of these projections has been characterized in mice, their ontogeny has not been studied. In this study, we used the DiI-based tract tracing approach to study the development of BNST projections innervating several hypothalamic areas including the PVH, DMH, ARH, and LHA. These results indicate that projections from the dorsomedial division of the anterior BNST to hypothalamic nuclei are immature at birth and substantially reach the PVH, DMH, and the LHA at P10. In the ARH, only sparse fibers are observed at P10, but their density increased markedly between P12 and P14. Collectively, these findings provide new insight into the ontogeny of hypothalamic circuits, and highlight the importance of considering the developmental context as a direct modulator in their proper formation.
Collapse
Affiliation(s)
- Marc Lanzillo
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Manon Gervais
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
32
|
Al-Massadi O, Dieguez C, Schneeberger M, López M, Schwaninger M, Prevot V, Nogueiras R. Multifaceted actions of melanin-concentrating hormone on mammalian energy homeostasis. Nat Rev Endocrinol 2021; 17:745-755. [PMID: 34608277 DOI: 10.1038/s41574-021-00559-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
Melanin-concentrating hormone (MCH) is a small cyclic peptide expressed in all mammals, mainly in the hypothalamus. MCH acts as a robust integrator of several physiological functions and has crucial roles in the regulation of sleep-wake rhythms, feeding behaviour and metabolism. MCH signalling has a very broad endocrine context and is involved in physiological functions and emotional states associated with metabolism, such as reproduction, anxiety, depression, sleep and circadian rhythms. MCH mediates its functions through two receptors (MCHR1 and MCHR2), of which only MCHR1 is common to all mammals. Owing to the wide variety of MCH downstream signalling pathways, MCHR1 agonists and antagonists have great potential as tools for the directed management of energy balance disorders and associated metabolic complications, and translational strategies using these compounds hold promise for the development of novel treatments for obesity. This Review provides an overview of the numerous roles of MCH in energy and glucose homeostasis, as well as in regulation of the mesolimbic dopaminergic circuits that encode the hedonic component of food intake.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
| | - Carlos Dieguez
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Miguel López
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S1172, EGID, Lille, France
| | - Ruben Nogueiras
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
33
|
Kobayashi Y, Tomoshige S, Imakado K, Sekino Y, Koganezawa N, Shirao T, Diniz GB, Miyamoto T, Saito Y. Ciliary GPCR-based transcriptome as a key regulator of cilia length control. FASEB Bioadv 2021; 3:744-767. [PMID: 34485842 PMCID: PMC8409570 DOI: 10.1096/fba.2021-00029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
The primary cilium is a plasma membrane-protruding sensory organelle that efficiently conveys signaling cascades in a highly ordered microenvironment. Its signaling is mediated, in part, by a limited set of GPCRs preferentially enriched in the cilium membrane. This includes melanin-concentrating hormone (MCH) receptor 1 (MCHR1), which plays a role in feeding and mood. In addition to its receptor composition, the length of the cilium is a characteristic parameter that is implicated in its function. We previously found that MCH can dynamically shorten cilia length via the Gi/o and Akt pathways in both MCHR1-expressing hTERT-RPE1 cells (hRPE1 cells) and rat hippocampal neurons. However, the detailed mechanisms by which MCH regulates cilia length through ciliary MCHR1 remains unclear. In this study, we aimed to determine the transcriptome changes in MCHR1-expressing hRPE1 cells in response to MCH to identify the target molecules involved in cilia length regulation via MCHR1 activation. RNA sequencing analysis of ciliated cells subjected to MCH treatment showed upregulation of 424 genes and downregulation of 112 genes compared with static control cells. Validation by quantitative real-time PCR, knocking down, and CRISPR/Cas9-mediated knockout technology identified a molecule, PDZ and LIM domain-containing protein 5 (PDLIM5). Thus, it was considered as the most significant key factor for MCHR1-mediated shortening of cilia length. Additional analyses revealed that the actin-binding protein alpha-actinin 1/4 is a crucial downstream target of the PDLIM5 signaling pathway that exerts an effect on MCHR1-induced cilia shortening. In the endogenous MCHR1-expressing hippocampus, transcriptional upregulation of PDLIM5 and actinin 1/4, following the application of MCH, was detected when the MCHR1-positive cilia were shortened. Together, our transcriptome study based on ciliary MCHR1 function uncovered a novel and important regulatory step underlying cilia length control. These results will potentially serve as a basis for understanding the mechanism underlying the development of obesity and mood disorders.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| | - Sakura Tomoshige
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| | - Kosuke Imakado
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| | - Yuko Sekino
- Endowed Laboratory of Human Cell‐Based Drug DiscoveryGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Noriko Koganezawa
- Department of Neurobiology and BehaviorGraduate School of MedicineGunma UniversityMaebashiJapan
| | - Tomoaki Shirao
- Department of Neurobiology and BehaviorGraduate School of MedicineGunma UniversityMaebashiJapan
- AlzMed, Inc.TokyoJapan
| | - Giovanne B. Diniz
- California National Primate Research CenterUniversity of CaliforniaDavisCAUSA
| | - Tatsuo Miyamoto
- Department of Genetics and Cell BiologyResearch Institute for Radiation Biology and MedicineHiroshima UniversityHiroshimaJapan
| | - Yumiko Saito
- Graduate School of Integrated Sciences for LifeHiroshima UniversityHiroshimaJapan
| |
Collapse
|
34
|
Jasso KR, Kamba TK, Zimmerman AD, Bansal R, Engle SE, Everett T, Wu CH, Kulaga H, Reed RR, Berbari NF, McIntyre JC. An N-terminal fusion allele to study melanin concentrating hormone receptor 1. Genesis 2021; 59:e23438. [PMID: 34124835 DOI: 10.1002/dvg.23438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 11/10/2022]
Abstract
Cilia on neurons play critical roles in both the development and function of the central nervous system (CNS). While it remains challenging to elucidate the precise roles for neuronal cilia, it is clear that a subset of G-protein-coupled receptors (GPCRs) preferentially localize to the cilia membrane. Further, ciliary GPCR signaling has been implicated in regulating a variety of behaviors. Melanin concentrating hormone receptor 1 (MCHR1), is a GPCR expressed centrally in rodents known to be enriched in cilia. Here we have used MCHR1 as a model ciliary GPCR to develop a strategy to fluorescently tag receptors expressed from the endogenous locus in vivo. Using CRISPR/Cas9, we inserted the coding sequence of the fluorescent protein mCherry into the N-terminus of Mchr1. Analysis of the fusion protein (mCherry MCHR1) revealed its localization to neuronal cilia in the CNS, across multiple developmental time points and in various regions of the adult brain. Our approach simultaneously produced fortuitous in/dels altering the Mchr1 start codon resulting in a new MCHR1 knockout line. Functional studies using electrophysiology show a significant alteration of synaptic strength in MCHR1 knockout mice. A reduction in strength is also detected in mice homozygous for the mCherry insertion, suggesting that while the strategy is useful for monitoring the receptor, activity could be altered. However, both lines should aid in studies of MCHR1 function and contribute to our understanding of MCHR1 signaling in the brain. Additionally, this approach could be expanded to aid in the study of other ciliary GPCRs.
Collapse
Affiliation(s)
- Kalene R Jasso
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA.,Graduate Program in Biomedical Sciences, Neuroscience Concentration, University of Florida, Gainesville, Florida, USA
| | - Tisianna K Kamba
- Graduate Program in Biomedical Sciences, Neuroscience Concentration, University of Florida, Gainesville, Florida, USA
| | - Arthur D Zimmerman
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Ruchi Bansal
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Staci E Engle
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Thomas Everett
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Chang-Hung Wu
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Heather Kulaga
- Department of Molecular Genetics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Randal R Reed
- Department of Molecular Genetics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicolas F Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Jeremy C McIntyre
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
35
|
Izawa S, Yoneshiro T, Kondoh K, Nakagiri S, Okamatsu-Ogura Y, Terao A, Minokoshi Y, Yamanaka A, Kimura K. Melanin-concentrating hormone-producing neurons in the hypothalamus regulate brown adipose tissue and thus contribute to energy expenditure. J Physiol 2021; 600:815-827. [PMID: 33899241 DOI: 10.1113/jp281241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/20/2021] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS Melanin-concentrating hormone (MCH) neuron-ablated mice exhibit increased energy expenditure and reduced fat weight. Increased brown adipose tissue (BAT) activity and locomotor activity-independent energy expenditure contributed to body weight reduction in MCH neuron-ablated mice. MCH neurons send inhibitory input to the medullary raphe nucleus to modulate BAT activity. ABSTRACT Hypothalamic melanin-concentrating hormone (MCH) peptide robustly affects energy homeostasis. However, it is unclear whether and how MCH-producing neurons, which contain and release a variety of neuropeptides/transmitters, regulate energy expenditure in the central nervous system and peripheral tissues. We thus examined the regulation of energy expenditure by MCH neurons, focusing on interscapular brown adipose tissue (BAT) activity. MCH neuron-ablated mice exhibited reduced body weight, increased oxygen consumption, and increased BAT activity, which improved locomotor activity-independent energy expenditure. Trans-neuronal retrograde tracing with the recombinant pseudorabies virus revealed that MCH neurons innervate BAT via the sympathetic premotor region in the medullary raphe nucleus (MRN). MRN neurons were activated by MCH neuron ablation. Therefore, endogenous MCH neuron activity negatively modulates energy expenditure via BAT inhibition. MRN neurons might receive inhibitory input from MCH neurons to suppress BAT activity.
Collapse
Affiliation(s)
- Shuntaro Izawa
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.,JSPS Research Fellowship for Young Scientists, Tokyo, 102-0083, Japan
| | - Takeshi Yoneshiro
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, 153-8904, Japan
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8585, Japan
| | - Shohei Nakagiri
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Akira Terao
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Department of Biology, School of Biological Sciences, Tokai University, Sapporo, 005-8601, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8585, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| |
Collapse
|
36
|
Barbier M, González JA, Houdayer C, Burdakov D, Risold P, Croizier S. Projections from the dorsomedial division of the bed nucleus of the stria terminalis to hypothalamic nuclei in the mouse. J Comp Neurol 2021; 529:929-956. [PMID: 32678476 PMCID: PMC7891577 DOI: 10.1002/cne.24988] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
As stressful environment is a potent modulator of feeding, we seek in the present work to decipher the neuroanatomical basis for an interplay between stress and feeding behaviors. For this, we combined anterograde and retrograde tracing with immunohistochemical approaches to investigate the patterns of projections between the dorsomedial division of the bed nucleus of the stria terminalis (BNST), well connected to the amygdala, and hypothalamic structures such as the paraventricular (PVH) and dorsomedial (DMH), the arcuate (ARH) nuclei and the lateral hypothalamic areas (LHA) known to control feeding and motivated behaviors. We particularly focused our study on afferences to proopiomelanocortin (POMC), agouti-related peptide (AgRP), melanin-concentrating-hormone (MCH) and orexin (ORX) neurons characteristics of the ARH and the LHA, respectively. We found light to intense innervation of all these hypothalamic nuclei. We particularly showed an innervation of POMC, AgRP, MCH and ORX neurons by the dorsomedial and dorsolateral divisions of the BNST. Therefore, these results lay the foundation for a better understanding of the neuroanatomical basis of the stress-related feeding behaviors.
Collapse
Affiliation(s)
- Marie Barbier
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
- Department of PsychiatrySeaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - J. Antonio González
- The Francis Crick InstituteLondonUK
- The Rowett Institute, School of MedicineMedical Sciences and Nutrition, University of AberdeenAberdeenUK
| | - Christophe Houdayer
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
| | - Denis Burdakov
- The Francis Crick InstituteLondonUK
- Neurobehavioural Dynamics Lab, Institute for Neuroscience, D‐HESTSwiss Federal Institute of Technology / ETH ZürichZürichSwitzerland
| | - Pierre‐Yves Risold
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
| | - Sophie Croizier
- University of LausanneCenter for Integrative GenomicsLausanneSwitzerland
| |
Collapse
|
37
|
Lord MN, Subramanian K, Kanoski SE, Noble EE. Melanin-concentrating hormone and food intake control: Sites of action, peptide interactions, and appetition. Peptides 2021; 137:170476. [PMID: 33370567 PMCID: PMC8025943 DOI: 10.1016/j.peptides.2020.170476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
Given the increased prevalence of obesity and its associated comorbidities, understanding the mechanisms through which the brain regulates energy balance is of critical importance. The neuropeptide melanin-concentrating hormone (MCH) is produced in the lateral hypothalamic area and the adjacent incerto-hypothalamic area and promotes both food intake and energy conservation, overall contributing to body weight gain. Decades of research into this system has provided insight into the neural pathways and mechanisms (behavioral and neurobiological) through which MCH stimulates food intake. Recent technological advancements that allow for selective manipulation of MCH neuron activity have elucidated novel mechanisms of action for the hyperphagic effects of MCH, implicating neural "volume" transmission in the cerebrospinal fluid and sex-specific effects of MCH on food intake control as understudied areas for future investigation. Highlighted here are historical and recent findings that illuminate the neurobiological mechanisms through which MCH promotes food intake, including the identification of various specific neural signaling pathways and interactions with other peptide systems. We conclude with a framework that the hyperphagic effects of MCH signaling are predominantly mediated through enhancement of an "appetition" process in which early postoral prandial signals promote further caloric consumption.
Collapse
Affiliation(s)
- Magen N Lord
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30606, USA
| | - Keshav Subramanian
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA; Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30606, USA.
| |
Collapse
|
38
|
Gómez-Martínez DG, Ramos M, del Valle-Padilla JL, Rosales JH, Robles F, Ramos F. A bioinspired model of short-term satiety of hunger influenced by food properties in virtual creatures. COGN SYST RES 2021. [DOI: 10.1016/j.cogsys.2020.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
39
|
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
|
40
|
Kobayashi Y, Okada T, Miki D, Sekino Y, Koganezawa N, Shirao T, Diniz GB, Saito Y. Properties of primary cilia in melanin-concentrating hormone receptor 1-bearing hippocampal neurons in vivo and in vitro. Neurochem Int 2020; 142:104902. [PMID: 33197527 DOI: 10.1016/j.neuint.2020.104902] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/31/2022]
Abstract
The primary cilium is a solitary organelle that organizes a sensitive signaling hub in a highly ordered microenvironment. Cilia are plastic structures, changing their length in response to bioactive substances, and ciliary length may be regulated to ensure efficient signaling capacity. Mammalian brain neurons possess primary cilia that are enriched in a set of G protein-coupled receptors (GPCRs), including the feeding-related melanin-concentrating hormone (MCH) receptor 1 (MCHR1). We previously demonstrated a novel biological phenomenon, ciliary MCHR1-mediated cilia length shortening through Gi/o and Akt signaling, using a simple cell culture model of human retinal pigmented epithelial RPE1 cells exogenously expressing MCHR1. In the present study, we characterized the properties of endogenous MCHR1-expressing primary cilia in hippocampal neurons in rodents. Using cultured dissociated rat hippocampal neurons in vitro, we showed that MCH triggered cilia length reduction involved in MCHR1-Gi/o and -Akt signaling. In rat hippocampal slice cultures with preservation of the cytoarchitecture and cell populations, ciliary MCHR1 was abundantly located in the CA1 and CA3 regions, but not in the dentate gyrus. Notably, treatment of slice cultures with MCH induced Gi/o- and Akt-dependent cilia shortening in the CA1 region without influencing cilia length in the CA3 region. Regarding the in vivo mouse brain, we observed higher levels of ciliary MCHR1 in the CA1 and CA3 regions as well as in slice cultures. In the starved state mice, a marked increase in MCH mRNA expression was detected in the lateral hypothalamus. Furthermore, MCHR1-positive cilia length in the hippocampal CA1 region was significantly shortened in fasted mice compared with fed mice. The present findings focused on the hippocampus provide a potential approach to investigate how MCHR1-driven cilia shortening regulates neuronal activity and physiological function toward feeding and memory tasks.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Tomoya Okada
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Daisuke Miki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Yuko Sekino
- Endowed Laboratory of Human Cell-Based Drug Discovery, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan; AlzMed,Inc., UT South Clinical Research Building, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8485, Japan
| | - Giovanne B Diniz
- Department of Neurosurgery, Yale School of Medicine, 310 Cedar St, New Haven, CT, 06520, USA
| | - Yumiko Saito
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan.
| |
Collapse
|
41
|
Kobayashi Y, Hamamoto A, Saito Y. Analysis of ciliary status via G-protein-coupled receptors localized on primary cilia. Microscopy (Oxf) 2020; 69:277-285. [PMID: 32627821 DOI: 10.1093/jmicro/dfaa035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/20/2020] [Accepted: 07/02/2020] [Indexed: 11/14/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) comprise the largest and most diverse cell surface receptor family, with more than 800 known GPCRs identified in the human genome. Binding of an extracellular cue to a GPCR results in intracellular G protein activation, after which a sequence of events, can be amplified and optimized by selective binding partners and downstream effectors in spatially discrete cellular environments. Because GPCRs are widely expressed in the body, they help to regulate an incredible range of physiological processes from sensation to growth to hormone responses. Indeed, it is estimated that ∼ 30% of all clinically approved drugs act by binding to GPCRs. The primary cilium is a sensory organelle composed of a microtubule axoneme that extends from the basal body. The ciliary membrane is highly enriched in specific signaling components, allowing the primary cilium to efficiently convey signaling cascades in a highly ordered microenvironment. Recent data demonstrated that a limited number of non-olfactory GPCRs, including somatostatin receptor 3 and melanin-concentrating hormone receptor 1 (MCHR1), are selectively localized to cilia on several mammalian cell types including neuronal cells. Utilizing cilia-specific cell biological and molecular biological approaches, evidence has accumulated to support the biological importance of ciliary GPCR signaling followed by cilia structural changes. Thus, cilia are now considered a unique sensory platform for integration of GPCR signaling toward juxtaposed cytoplasmic structures. Herein, we review ciliary GPCRs and focus on a novel role of MCHR1 in ciliary length control that will impact ciliary signaling capacity and neuronal function.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8521, Japan
| | - Akie Hamamoto
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, Gifu 502-0857, Japan
| | - Yumiko Saito
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8521, Japan
| |
Collapse
|
42
|
Bandaru SS, Khanday MA, Ibrahim N, Naganuma F, Vetrivelan R. Sleep-Wake Control by Melanin-Concentrating Hormone (MCH) Neurons: a Review of Recent Findings. Curr Neurol Neurosci Rep 2020; 20:55. [PMID: 33006677 DOI: 10.1007/s11910-020-01075-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE OF THE REVIEW Melanin-concentrating hormone (MCH)-expressing neurons located in the lateral hypothalamus are considered as an integral component of sleep-wake circuitry. However, the precise role of MCH neurons in sleep-wake regulation has remained unclear, despite several years of research employing a wide range of techniques. We review recent data on this aspect, which are mostly inconsistent, and propose a novel role for MCH neurons in sleep regulation. RECENT FINDINGS While almost all studies using "gain-of-function" approaches show an increase in rapid eye movement sleep (or paradoxical sleep; PS), loss-of-function approaches have not shown reductions in PS. Similarly, the reported changes in wakefulness or non-rapid eye movement sleep (slow-wave sleep; SWS) with manipulation of the MCH system using conditional genetic methods are inconsistent. Currently available data do not support a role for MCH neurons in spontaneous sleep-wake but imply a crucial role for them in orchestrating sleep-wake responses to changes in external and internal environments.
Collapse
Affiliation(s)
- Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
| | - Mudasir A Khanday
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Nazifa Ibrahim
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA.,Department of Public Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA.,Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA. .,Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Phan J, Alhassen L, Argelagos A, Alhassen W, Vachirakorntong B, Lin Z, Sanathara N, Alachkar A. Mating and parenting experiences sculpture mood-modulating effects of oxytocin-MCH signaling. Sci Rep 2020; 10:13611. [PMID: 32788646 PMCID: PMC7423941 DOI: 10.1038/s41598-020-70667-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/31/2020] [Indexed: 11/09/2022] Open
Abstract
The two hypothalamic neuropeptides oxytocin and melanin concentrating hormone (MCH) share several physiological actions such as the control of maternal care, sexual behavior, and emotions. In this study, we uncover the role for the oxytocin-MCH signaling pathway in mood regulation. We identify discrete effects of oxytocin-MCH signaling on depressive behavior and demonstrate that parenting and mating experiences shape these effects. We show that the selective deletion of OXT receptors from MCH neurons increases and decreases depressive behavior in sexually naïve and late postpartum female mice respectively, with no effect on sexually naïve male mice. We demonstrate that both parenting experience and mood-regulating effects of oxytocin-MCH are associated with synaptic plasticity in the reward and fear circuits revealed by the alterations of Arc expressions, which are associated with the depressive behavior. Finally, we uncover the sex-dependent effects of mating on depressive behavior; while the sexual activity reduces the basal levels of depressive behavior in male mice, it reduces in female mice evoked-depression only. We demonstrate that the oxytocin-MCH pathway mediates the effects of sexual activity on depressive behavior. Our data suggest that the oxytocin-MCH pathway can serve as a potential therapeutic target for the treatment of major depression and postpartum mood disorders.
Collapse
Affiliation(s)
- Joseph Phan
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Lamees Alhassen
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Allan Argelagos
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Wedad Alhassen
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Benjamin Vachirakorntong
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Zitong Lin
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Nayna Sanathara
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, University of California, Irvine, 356A Med Surge II, Irvine, CA, 92697-4625, USA. .,Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
44
|
Ong ZY, McNally GP. CART in energy balance and drug addiction: Current insights and mechanisms. Brain Res 2020; 1740:146852. [DOI: 10.1016/j.brainres.2020.146852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022]
|
45
|
Battagello DS, Lorenzon AR, Diniz GB, Motta-Teixeira LC, Klein MO, Ferreira JGP, Arias CM, Adamantidis A, Sita LV, Cipolla-Neto J, Bevilacqua EMAF, Sawchenko PE, Bittencourt JC. The Rat Mammary Gland as a Novel Site of Expression of Melanin-Concentrating Hormone Receptor 1 mRNA and Its Protein Immunoreactivity. Front Endocrinol (Lausanne) 2020; 11:463. [PMID: 32849267 PMCID: PMC7411258 DOI: 10.3389/fendo.2020.00463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/12/2020] [Indexed: 11/24/2022] Open
Abstract
Lactation is a complex physiological process, depending on orchestrated central and peripheral events, including substantial brain plasticity. Among these events is a novel expression of pro-melanin-concentrating hormone (Pmch) mRNA in the rodent hypothalamus, such as the ventral part of the medial preoptic area (vmMPOA). This expression reaches its highest levels around postpartum day 19 (PPD19), when dams transition from lactation to the weaning period. The appearance of this lactation-related Pmch expression occurs simultaneously with the presence of one of the Pmch products, melanin-concentrating hormone (MCH), in the serum. Given the relevance of the MPOA to maternal physiology and the contemporaneity between Pmch expression in this structure and the weaning period, we hypothesized that MCH has a role in the termination of lactation, acting as a mediator between central and peripheral changes. To test this, we investigated the presence of the MCH receptor 1 (MCHR1) and its gene expression in the mammary gland of female rats in different stages of the reproductive cycle. To that end, in situ hybridization, RT-PCR, RT-qPCR, nucleotide sequencing, immunohistochemistry, and Western blotting were employed. Although Mchr1 expression was detected in the epidermis and dermis of both diestrus and lactating rats, parenchymal expression was exclusively found in the functional mammary gland of lactating rats. The expression of Mchr1 mRNA oscillated through the lactation period and reached its maximum in PPD19 dams. Presence of MCHR1 was confirmed with immunohistochemistry with preferential location of MCHR1 immunoreactive cells in the alveolar secretory cells. As was the case for gene expression, the MCHR1 protein levels were significantly higher in PPD19 than in other groups. Our data demonstrate the presence of an anatomical basis for the participation of MCH peptidergic system on the control of lactation through the mammary gland, suggesting that MCH could modulate a prolactation action in early postpartum days and the opposite role at the end of the lactation.
Collapse
Affiliation(s)
- Daniella S. Battagello
- Instituto de Psicologia, Nucleo de Neurociencias e Comportamento, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Universidade de São Paulo, São Paulo, Brazil
| | - Aline R. Lorenzon
- Departmento de Biologia Celular e Do Desenvolvimento, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Giovanne B. Diniz
- Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Universidade de São Paulo, São Paulo, Brazil
| | - Lívia C. Motta-Teixeira
- Departmento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Marianne O. Klein
- Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Universidade de São Paulo, São Paulo, Brazil
| | - Jozélia G. P. Ferreira
- Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Universidade de São Paulo, São Paulo, Brazil
| | - Carlos M. Arias
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | | | - Luciane V. Sita
- Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Universidade de São Paulo, São Paulo, Brazil
| | - José Cipolla-Neto
- Departmento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Estela M. A. F. Bevilacqua
- Departmento de Biologia Celular e Do Desenvolvimento, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Paul E. Sawchenko
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Jackson C. Bittencourt
- Instituto de Psicologia, Nucleo de Neurociencias e Comportamento, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Cheong RY, Gabery S, Petersén Å. The Role of Hypothalamic Pathology for Non-Motor Features of Huntington's Disease. J Huntingtons Dis 2020; 8:375-391. [PMID: 31594240 PMCID: PMC6839491 DOI: 10.3233/jhd-190372] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Huntington’s disease (HD) is a fatal genetic neurodegenerative disorder. It has mainly been considered a movement disorder with cognitive symptoms and these features have been associated with pathology of the striatum and cerebral cortex. Importantly, individuals with the mutant huntingtin gene suffer from a spectrum of non-motor features often decades before the motor disorder manifests. These symptoms and signs include a range of psychiatric symptoms, sleep problems and metabolic changes with weight loss particularly in later stages. A higher body mass index at diagnosis is associated with slower disease progression. The common psychiatric symptom of apathy progresses with the disease. The fact that non-motor features are present early in the disease and that they show an association to disease progression suggest that unravelling the underlying neurobiological mechanisms may uncover novel targets for early disease intervention and better symptomatic treatment. The hypothalamus and the limbic system are important brain regions that regulate emotion, social cognition, sleep and metabolism. A number of studies using neuroimaging, postmortem human tissue and genetic manipulation in animal models of the disease has collectively shown that the hypothalamus and the limbic system are affected in HD. These findings include the loss of neuropeptide-expressing neurons such as orexin (hypocretin), oxytocin, vasopressin, somatostatin and VIP, and increased levels of SIRT1 in distinct nuclei of the hypothalamus. This review provides a summary of the results obtained so far and highlights the potential importance of these changes for the understanding of non-motor features in HD.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sanaz Gabery
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
47
|
Chang GQ, Karatayev O, Boorgu DSSK, Leibowitz SF. CCL2/CCR2 system in neuroepithelial radial glia progenitor cells: involvement in stimulatory, sexually dimorphic effects of maternal ethanol on embryonic development of hypothalamic peptide neurons. J Neuroinflammation 2020; 17:207. [PMID: 32650794 PMCID: PMC7353676 DOI: 10.1186/s12974-020-01875-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/16/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Clinical and animal studies show that alcohol consumption during pregnancy produces lasting behavioral disturbances in offspring, including increased alcohol drinking, which are linked to inflammation in the brain and disturbances in neurochemical systems that promote these behaviors. These include the neuropeptide, melanin-concentrating hormone (MCH), which is mostly expressed in the lateral hypothalamus (LH). Maternal ethanol administration at low-to-moderate doses, while stimulating MCH neurons without affecting apoptosis or gliogenesis, increases in LH the density of neurons expressing the inflammatory chemokine C-C motif ligand 2 (CCL2) and its receptor CCR2 and their colocalization with MCH. These neural effects associated with behavioral changes are reproduced by maternal CCL2 administration, reversed by a CCR2 antagonist, and consistently stronger in females than males. The present study investigates in the embryo the developmental origins of this CCL2/CCR2-mediated stimulatory effect of maternal ethanol exposure on MCH neurons. METHODS Pregnant rats from embryonic day 10 (E10) to E15 during peak neurogenesis were orally administered ethanol at a moderate dose (2 g/kg/day) or peripherally injected with CCL2 or CCR2 antagonist to test this neuroimmune system's role in ethanol's actions. Using real-time quantitative PCR, immunofluorescence histochemistry, in situ hybridization, and confocal microscopy, we examined in embryos at E19 the CCL2/CCR2 system and MCH neurons in relation to radial glia progenitor cells in the hypothalamic neuroepithelium where neurons are born and radial glia processes projecting laterally through the medial hypothalamus that provide scaffolds for neuronal migration into LH. RESULTS We demonstrate that maternal ethanol increases radial glia cell density and their processes while stimulating the CCL2/CCR2 system and these effects are mimicked by maternal administration of CCL2 and blocked by a CCR2 antagonist. While stimulating CCL2 colocalization with radial glia and neurons but not microglia, ethanol increases MCH neuronal number near radial glia cells and making contact along their processes projecting into LH. Further tests identify the CCL2/CCR2 system in NEP as a primary source of ethanol's sexually dimorphic actions. CONCLUSIONS These findings provide new evidence for how an inflammatory chemokine pathway functions within neuroprogenitor cells to mediate ethanol's long-lasting, stimulatory effects on peptide neurons linked to adolescent drinking behavior.
Collapse
Affiliation(s)
- Guo-Qing Chang
- The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Olga Karatayev
- The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | | | | |
Collapse
|
48
|
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: 2.2] [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
|
49
|
MCH Neurons Regulate Permeability of the Median Eminence Barrier. Neuron 2020; 107:306-319.e9. [PMID: 32407670 PMCID: PMC7383232 DOI: 10.1016/j.neuron.2020.04.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 03/06/2020] [Accepted: 04/20/2020] [Indexed: 01/17/2023]
Abstract
Melanin-concentrating hormone (MCH)-expressing neurons are key regulators of energy and glucose homeostasis. Here, we demonstrate that they provide dense projections to the median eminence (ME) in close proximity to tanycytes and fenestrated vessels. Chemogenetic activation of MCH neurons as well as optogenetic stimulation of their projections in the ME enhance permeability of the ME by increasing fenestrated vascular loops and enhance leptin action in the arcuate nucleus of the hypothalamus (ARC). Unbiased phosphoRiboTrap-based assessment of cell activation upon chemogenetic MCH neuron activation reveals MCH-neuron-dependent regulation of endothelial cells. MCH neurons express the vascular endothelial growth factor A (VEGFA), and blocking VEGF-R signaling attenuates the leptin-sensitizing effect of MCH neuron activation. Our experiments reveal that MCH neurons directly regulate permeability of the ME barrier, linking the activity of energy state and sleep regulatory neurons to the regulation of hormone accessibility to the ARC. MCH neurons provide dense projections to the median eminence MCH neuron activation promotes permeability of the median eminence barrier MCH neuron activation enhances microvessel fenestration in the ME MCH neuron activation enhances leptin action in the arcuate nucleus
Collapse
|
50
|
Morganstern I, Gulati G, Leibowitz SF. Role of melanin-concentrating hormone in drug use disorders. Brain Res 2020; 1741:146872. [PMID: 32360868 DOI: 10.1016/j.brainres.2020.146872] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 04/17/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022]
Abstract
Melanin-concentrating hormone (MCH) is a neuropeptide primarily transcribed in the lateral hypothalamus (LH), with vast projections to many areas throughout the central nervous system that play an important role in motivated behaviors and drug use. Anatomical, pharmacological and genetic studies implicate MCH in mediating the intake and reinforcement of commonly abused substances, acting by influencing several systems including the mesolimbic dopaminergic system, glutamatergic as well as GABAergic signaling and being modulated by inflammatory neuroimmune pathways. Further support for the role of MCH in controlling behavior related to drug use will be discussed as it relates to cerebral ventricular volume transmission and intracellular molecules including cocaine- and amphetamine-regulated transcript peptide, dopamine- and cAMP-regulated phosphoprotein 32 kDa. The primary goal of this review is to introduce and summarize current literature surrounding the role of MCH in mediating the intake and reinforcement of commonly abused drugs, such as alcohol, cocaine, amphetamine, nicotine and opiates.
Collapse
Affiliation(s)
| | - Gazal Gulati
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|