1
|
Bjorness TE, Greene RW. Orexin-mediated motivated arousal and reward seeking. Peptides 2024; 180:171280. [PMID: 39159833 DOI: 10.1016/j.peptides.2024.171280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
2
|
Wang X, Wu X, Wu H, Xiao H, Hao S, Wang B, Li C, Bleymehl K, Kauschke SG, Mack V, Ferger B, Klein H, Zheng R, Duan S, Wang H. Neural adaption in midbrain GABAergic cells contributes to high-fat diet-induced obesity. SCIENCE ADVANCES 2023; 9:eadh2884. [PMID: 37910621 PMCID: PMC10619925 DOI: 10.1126/sciadv.adh2884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023]
Abstract
Overeating disorders largely contribute to worldwide incidences of obesity. Available treatments are limited. Here, we discovered that long-term chemogenetic activation of ventrolateral periaqueductal gray (vlPAG) GABAergic cells rescue obesity of high-fat diet-induced obesity (DIO) mice. This was associated with the recovery of enhanced mIPSCs, decreased food intake, increased energy expenditure, and inguinal white adipose tissue (iWAT) browning. In vivo calcium imaging confirmed vlPAG GABAergic suppression for DIO mice, with corresponding reduction in intrinsic excitability. Single-nucleus RNA sequencing identified transcriptional expression changes in GABAergic cell subtypes in DIO mice, highlighting Cacna2d1 as of potential importance. Overexpressing CACNA2D1 in vlPAG GABAergic cells of DIO mice rescued enhanced mIPSCs and calcium response, reversed obesity, and therefore presented here as a potential target for obesity treatment.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaotong Wu
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Wu
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang 310058, China
| | - Hanyang Xiao
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Sijia Hao
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100091, China
| | - Chen Li
- Department of Human Genetics and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Katherin Bleymehl
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Stefan G. Kauschke
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Volker Mack
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Boris Ferger
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Holger Klein
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100091, China
| | - Shumin Duan
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Wang
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Lingang Laboratory, Shanghai 200031, China
| |
Collapse
|
3
|
Bouâouda H, Jha PK. Orexin and MCH neurons: regulators of sleep and metabolism. Front Neurosci 2023; 17:1230428. [PMID: 37674517 PMCID: PMC10478345 DOI: 10.3389/fnins.2023.1230428] [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: 05/28/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Sleep-wake and fasting-feeding are tightly coupled behavioral states that require coordination between several brain regions. The mammalian lateral hypothalamus (LH) is a functionally and anatomically complex brain region harboring heterogeneous cell populations that regulate sleep, feeding, and energy metabolism. Significant attempts were made to understand the cellular and circuit bases of LH actions. Rapid advancements in genetic and electrophysiological manipulation help to understand the role of discrete LH cell populations. The opposing action of LH orexin/hypocretin and melanin-concentrating hormone (MCH) neurons on metabolic sensing and sleep-wake regulation make them the candidate to explore in detail. This review surveys the molecular, genetic, and neuronal components of orexin and MCH signaling in the regulation of sleep and metabolism.
Collapse
Affiliation(s)
- Hanan Bouâouda
- Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Pawan Kumar Jha
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
4
|
Chatterjee O, Gopalakrishnan L, Pullimamidi D, Raj C, Yelamanchi S, Gangadharappa BS, Nair B, Mahadevan A, Raju R, Keshava Prasad TS. A molecular network map of orexin-orexin receptor signaling system. J Cell Commun Signal 2023; 17:217-227. [PMID: 36480100 PMCID: PMC10030760 DOI: 10.1007/s12079-022-00700-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/04/2022] [Accepted: 09/15/2022] [Indexed: 12/13/2022] Open
Abstract
Orexins are excitatory neuropeptides, which are predominantly associated with feeding behavior, sleep-wake cycle and energy homeostasis. The orexinergic system comprises of HCRTR1 and HCRTR2, G-protein-coupled receptors of rhodopsin family and the endogenous ligands processed from HCRT pro-hormone, Orexin A and Orexin B. These neuropeptides are biosynthesized by the orexin neurons present in the lateral hypothalamus area, with dense projections to other brain regions. The orexin-receptor signaling is implicated in various metabolic as well as neurological disorders, making it a promising target for pharmacological interventions. However, there is limited information available on the collective representation of the signal transduction pathways pertaining to the orexin-orexin receptor signaling system. Here, we depict a compendium of the Orexin A/B stimulated reactions in the form of a basic signaling pathway map. This map catalogs the reactions into five categories: molecular association, activation/inhibition, catalysis, transport, and gene regulation. A total of 318 downstream molecules were annotated adhering to the guidelines of NetPath curation. This pathway map can be utilized for further assessment of signaling events associated with orexin-mediated physiological functions and is freely available on WikiPathways, an open-source pathway database ( https://www.wikipathways.org/index.php/Pathway:WP5094 ).
Collapse
Affiliation(s)
- Oishi Chatterjee
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, 690 525, Kollam, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India
| | - Lathika Gopalakrishnan
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India
- Manipal Academy of Higher Education (MAHE), 576 104, Manipal, India
| | | | - Chinmayi Raj
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
| | - Soujanya Yelamanchi
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
| | | | - Bipin Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, 690 525, Kollam, India
| | - Anita Mahadevan
- Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 560 029, Bangalore, India
- Department of Neuropathology, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 560 029, Bangalore, India
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India.
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India.
| |
Collapse
|
5
|
Villano I, La Marra M, Di Maio G, Monda V, Chieffi S, Guatteo E, Messina G, Moscatelli F, Monda M, Messina A. Physiological Role of Orexinergic System for Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8353. [PMID: 35886210 PMCID: PMC9323672 DOI: 10.3390/ijerph19148353] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023]
Abstract
Orexins, or hypocretins, are excitatory neuropeptides involved in the regulation of feeding behavior and the sleep and wakefulness states. Since their discovery, several lines of evidence have highlighted that orexin neurons regulate a great range of physiological functions, giving it the definition of a multitasking system. In the present review, we firstly describe the mechanisms underlining the orexin system and their interactions with the central nervous system (CNS). Then, the system's involvement in goal-directed behaviors, sleep/wakefulness state regulation, feeding behavior and energy homeostasis, reward system, and aging and neurodegenerative diseases are described. Advanced evidence suggests that the orexin system is crucial for regulating many physiological functions and could represent a promising target for therapeutical approaches to obesity, drug addiction, and emotional stress.
Collapse
Affiliation(s)
- Ines Villano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Marco La Marra
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Girolamo Di Maio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Vincenzo Monda
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80138 Naples, Italy; (V.M.); (E.G.)
| | - Sergio Chieffi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Ezia Guatteo
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80138 Naples, Italy; (V.M.); (E.G.)
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (G.M.); (F.M.)
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (G.M.); (F.M.)
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Antonietta Messina
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| |
Collapse
|
6
|
Butler-Struben HM, Kentner AC, Trainor BC. What's wrong with my experiment?: The impact of hidden variables on neuropsychopharmacology research. Neuropsychopharmacology 2022; 47:1285-1291. [PMID: 35338255 PMCID: PMC9117327 DOI: 10.1038/s41386-022-01309-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/22/2022] [Accepted: 03/10/2022] [Indexed: 12/24/2022]
Abstract
The field of neuropsychopharmacology relies on behavioral assays to quantify behavioral processes related to mental illness and substance use disorders. Although these assays have been highly informative, sometimes laboratories have unpublished datasets from experiments that "didn't work". Often this is because expected outcomes were not observed in positive or negative control groups. While this can be due to experimenter error, an important alternative is that under-appreciated environmental factors can have a major impact on results. "Hidden variables" such as circadian cycles, husbandry, and social environments are often omitted in methods sections, even though there is a strong body of literature documenting their impact on physiological and behavioral outcomes. Applying this knowledge in a more critical manner could provide behavioral neuroscientists with tools to develop better testing methods, improve the external validity of behavioral techniques, and make better comparisons of experimental data across institutions. Here we review the potential impact of "hidden variables" that are commonly overlooked such as light-dark cycles, transport stress, cage ventilation, and social housing structure. While some of these conditions may not be under direct control of investigators, it does not diminish the potential impact of these variables on experimental results. We provide recommendations to investigators on which variables to report in publications and how to address "hidden variables" that impact their experimental results.
Collapse
Affiliation(s)
| | - Amanda C Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Brian C Trainor
- Animal Behavior Graduate Group, University of California, Davis, CA, 95616, USA.
- Department of Psychology, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
7
|
Agee LA, Nemchek V, Malone CA, Lee HJ, Monfils MH. Appetitive Behavior in the Social Transmission of Food Preference Paradigm Predicts Activation of Orexin-A producing Neurons in a Sex-Dependent Manner. Neuroscience 2022; 481:30-46. [PMID: 34843892 PMCID: PMC9246717 DOI: 10.1016/j.neuroscience.2021.11.032] [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: 06/12/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/22/2022]
Abstract
Orexin-producing cells in the lateral hypothalamic area have been shown to be involved in a wide variety of behavioral and cognitive functions, including the recall of appetitive associations and a variety of social behaviors. Here, we investigated the role of orexin in the acquisition and recall of socially transmitted food preferences in the rat. Rats were euthanized following either acquisition, short-term recall, or long-term recall of a socially transmitted food preference and their brains were processed for orexin-A and c-Fos expression. We found that while there were no significant differences in c-Fos expression between control and experimental subjects at any of the tested timepoints, females displayed significantly more activity in both orexinergic and non-orexinergic cells in the lateral hypothalamus. In the infralimbic cortex, we found that social behavior was significantly predictive of c-Fos expression, with social behaviors related to olfactory exploration appearing to be particularly influential. We additionally found that appetitive behavior was significantly predictive of orexin-A activity in a sex-dependent matter, with the total amount eaten correlating negatively with orexin-A/c-Fos colocalization in male rats but not female rats. These findings suggest a potential sex-specific role for the orexin system in balancing the stimulation of feeding behavior with the sleep/wake cycle.
Collapse
Affiliation(s)
- Laura A Agee
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Victoria Nemchek
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Cassidy A Malone
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Hongjoo J Lee
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Marie-H Monfils
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
8
|
Martins-Oliveira M, Tavares I, Goadsby PJ. Was it something I ate? Understanding the bidirectional interaction of migraine and appetite neural circuits. Brain Res 2021; 1770:147629. [PMID: 34428465 DOI: 10.1016/j.brainres.2021.147629] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/18/2022]
Abstract
Migraine attacks can involve changes of appetite: while fasting or skipping meals are often reported triggers in susceptible individuals, hunger or food craving are reported in the premonitory phase. Over the last decade, there has been a growing interest and recognition of the importance of studying these overlapping fields of neuroscience, which has led to novel findings. The data suggest additional studies are needed to unravel key neurobiological mechanisms underlying the bidirectional interaction between migraine and appetite. Herein, we review information about the metabolic migraine phenotype and explore migraine therapeutic targets that have a strong input on appetite neuronal circuits, including the calcitonin gene-related peptide (CGRP), the pituitary adenylate cyclase-activating polypeptide (PACAP) and the orexins. Furthermore, we focus on potential therapeutic peptide targets that are involved in regulation of feeding and play a role in migraine pathophysiology, such as neuropeptide Y, insulin, glucagon and leptin. We then examine the orexigenic - anorexigenic circuit feedback loop and explore glucose metabolism disturbances. Additionally, it is proposed a different perspective on the most reported feeding-related trigger - skipping meals - as well as a link between contrasting feeding behaviors (skipping meals vs food craving). Our review aims to increase awareness of migraine through the lens of appetite neurobiology in order to improve our understanding of the earlier phase of migraine, encourage better studies and cross-disciplinary collaborations, and provide novel migraine-specific therapeutic opportunities.
Collapse
Affiliation(s)
- Margarida Martins-Oliveira
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK; Nutrition and Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas de Lisboa, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal.
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Institute of Investigation and Innovation in Health (i3S), University of Porto, Portugal.
| | - Peter J Goadsby
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK; Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Han D, Shi Y, Han F. The effects of orexin-A and orexin receptors on anxiety- and depression-related behaviors in a male rat model of post-traumatic stress disorder. J Comp Neurol 2021; 530:592-606. [PMID: 34387361 DOI: 10.1002/cne.25231] [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: 01/01/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
Orexin neurons play an important role in stress-related mental disorders including post-traumatic stress disorder (PTSD). Anxiety- and depression-related symptoms commonly occur in combination with PTSD. However, the role of the orexin system in mediating alterations in these affective symptoms remains unclear. The medial prefrontal cortex (mPFC) is implicated in both cognitive and emotional processing. In the present study, we investigated anxiety- and depression-related behavioral changes using the elevated plus maze, the sucrose preference test, and the open field test in male rats with single prolonged stress (SPS) induced-PTSD. The expression of orexin-A in the hypothalamus and orexin receptors (OX1R and OX2R) in the mPFC was detected and quantified by immunohistochemistry, western blotting, and real-time polymerase chain reaction. We found that the SPS rats exhibited enhanced levels of anxiety, reduced exploratory activities, and anhedonia. Furthermore, SPS resulted in reductions in the expression of orexin-A in the hypothalamus and the increased the expression of OX1R in the mPFC. The intracerebroventricular administration of orexin-A alleviated behavioral changes in SPS rats and partly restored the increased levels of OX1R in the mPFC. These results suggest that the orexin system plays a role in the anxiety- and depression-related symptoms observed in PTSD.
Collapse
Affiliation(s)
- Dan Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, China.,Department of Neonatology, The First Hospital of China Medical University, Shenyang, China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, China
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, China
| |
Collapse
|
10
|
Vitale RM, Iannotti FA, Schiano Moriello A, Tunisi L, Piscitelli F, Savopoulos R, Cristino L, De Petrocellis L, Amodeo P, Gray R, Di Marzo V. Identification and Characterization of Cannabidiol as an OX1R Antagonist by Computational and In Vitro Functional Validation. Biomolecules 2021; 11:1134. [PMID: 34439801 PMCID: PMC8394412 DOI: 10.3390/biom11081134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/22/2022] Open
Abstract
The potential, multifaceted therapeutic profile of cannabidiol (CBD), a major constituent derived from the Cannabis sativa plant, covers a wide range of neurological and psychiatric disorders, ranging from anxiety to pediatric epilepsy and drug addiction. However, the molecular targets responsible for these effects have been only partially identified. In this view, the involvement of the orexin system, the key regulator in arousal and the sleep/wake cycle, and in motivation and reward processes, including drug addiction, prompted us to explore, using computational and experimental approaches, the possibility that CBD could act as a ligand of orexin receptors, orexin 1 receptor of type 1 (OX1R) and type 2 (OX2R). Ligand-binding assays showed that CBD is a selective ligand of OX1R in the low micromolar range (Ki 1.58 ± 0.2 μM) while in vitro functional assays, carried out by intracellular calcium imaging and mobilization assays, showed that CBD acts as an antagonist at this receptor. Finally, the putative binding mode of CBD has been inferred by molecular docking and molecular dynamics simulations and its selectivity toward the OX1R subtype rationalized at the molecular level. This study provides the first evidence that CBD acts as an OX1R antagonist, supporting its potential use in addictive disorders and/or body weight regulation.
Collapse
Affiliation(s)
- Rosa Maria Vitale
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
- Epitech Group SpA, Saccolongo, 35100 Padova, Italy
| | - Lea Tunisi
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80131 Naples, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
| | - Ranjev Savopoulos
- GW Research Ltd., Sovereign House, Vision Park, Histon, Cambridge CB24 9BZ, UK; (R.S.); (R.G.)
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
| | - Luciano De Petrocellis
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
| | - Roy Gray
- GW Research Ltd., Sovereign House, Vision Park, Histon, Cambridge CB24 9BZ, UK; (R.S.); (R.G.)
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (F.A.I.); (F.P.); (L.C.); (L.D.); (P.A.)
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (A.S.M.); (L.T.)
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC G1V 4G5, Canada
| |
Collapse
|
11
|
Muthmainah M, Gogos A, Sumithran P, Brown RM. Orexins (hypocretins): The intersection between homeostatic and hedonic feeding. J Neurochem 2021; 157:1473-1494. [PMID: 33608877 DOI: 10.1111/jnc.15328] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
Orexins are hypothalamic neuropeptides originally discovered to play a role in the regulation of feeding behaviour. The broad connections of orexin neurons to mesocorticolimbic circuitry suggest they may play a role in mediating reward-related behaviour beyond homeostatic feeding. Here, we review the role of orexin in a variety of eating-related behaviour, with a focus on reward and motivation, and the neural circuits driving these effects. One emerging finding is the involvement of orexins in hedonic and appetitive behaviour towards palatable food, in addition to their role in homeostatic feeding. This review discusses the brain circuitry and possible mechanisms underlying the role of orexins in these behaviours. Overall, there is a marked bias in the literature towards studies involving male subjects. As such, future work needs to be done to involve female subjects. In summary, orexins play an important role in driving motivation for high salient rewards such as highly palatable food and may serve as the intersection between homeostatic and hedonic feeding.
Collapse
Affiliation(s)
- Muthmainah Muthmainah
- The Florey Institute of Neuroscience and Mental Health, Mental Health Research Theme, Parkville, Melbourne, Vic., Australia.,The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Vic., Australia.,Department of Anatomy, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Andrea Gogos
- The Florey Institute of Neuroscience and Mental Health, Mental Health Research Theme, Parkville, Melbourne, Vic., Australia
| | - Priya Sumithran
- Department of Medicine (Austin), University of Melbourne, Heidelberg, Vic., Australia.,Department of Endocrinology, Austin Health, Heidelberg, Vic., Australia
| | - Robyn M Brown
- The Florey Institute of Neuroscience and Mental Health, Mental Health Research Theme, Parkville, Melbourne, Vic., Australia.,The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Vic., Australia
| |
Collapse
|
12
|
Williams DL, Coiduras II, Parise EM, Maske CB. Hindbrain orexin 1 receptors blunt intake suppression by gastrointestinal nutrients and cholecystokinin in male rats. Peptides 2020; 133:170351. [PMID: 32579900 PMCID: PMC7556705 DOI: 10.1016/j.peptides.2020.170351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/19/2020] [Accepted: 06/19/2020] [Indexed: 01/06/2023]
Abstract
Hypothalamic orexin neurons project to many brain areas, including hindbrain structures such as the nucleus of the solitary tract (NTS) and area postrema (AP), where orexin 1 receptors (OX1Rs) are expressed. Hindbrain administration of orexin-A increases feeding and meal size, and blockade of hindbrain OX1Rs with the selective antagonist SB334867 has the opposite effect. Here we asked whether hindbrain OX1R stimulation or blockade alter rats' sensitivity to gastrointestinal satiety signals. Rats received 4th intracerebroventricular (icv) injections of vehicle or orexin-A, at a dose with no effect on its own, prior to an intragastric (IG) infusion of saline or a satiating volume of Ensure. IG Ensure suppressed subsequent chow intake, but orexin-A pretreatment significantly attenuated this IG nutrient-induced satiety at 2 h into the dark phase. In a second experiment, rats received NTS injections of vehicle or orexin-A before intraperitoneal (IP) injection of vehicle or the satiation hormone cholecystokinin (CCK). NTS orexin-A pretreatment completely blocked the intake-suppressive effect of CCK on dark-phase chow intake. Finally, we investigated the role of endogenous hindbrain OX1R activation by pretreating rats with 4th-icv injection of vehicle or SB334867 followed by IG infusion of saline or Ensure just before a chocolate Ensure licking test session. IG nutrient infusion suppressed Ensure intake, and blockade of hindbrain OX1Rs significantly prolonged that intake-suppressive effect. We conclude that hindbrain OX1Rs are a mechanism though which hypothalamic orexin neurons can reduce animals' sensitivity to gastrointestinal nutrient load, allowing them to consume more food.
Collapse
Affiliation(s)
- Diana L Williams
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301 USA.
| | - Isabel I Coiduras
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301 USA
| | - Eric M Parise
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301 USA
| | - Calyn B Maske
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301 USA
| |
Collapse
|
13
|
Lee J, Raycraft L, Johnson AW. The dynamic regulation of appetitive behavior through lateral hypothalamic orexin and melanin concentrating hormone expressing cells. Physiol Behav 2020; 229:113234. [PMID: 33130035 DOI: 10.1016/j.physbeh.2020.113234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The lateral hypothalamic area (LHA) is a heterogeneous brain structure extensively studied for its potent role in regulating energy balance. The anatomical and molecular diversity of the LHA permits the orchestration of responses to energy sensing cues from the brain and periphery. Two of the primary cell populations within the LHA associated with integration of this information are Orexin (ORX) and Melanin Concentrating Hormone (MCH). While both of these non-overlapping populations exhibit orexigenic properties, the activities of these two systems support feeding behavior through contrasting mechanisms. We describe the anatomical and functional properties as well as interaction with other neuropeptides and brain reward and hedonic systems. Specific outputs relating to arousal, food seeking, feeding, and metabolism are coordinated through these mechanisms. We then discuss how both the ORX and MCH systems harmonize in a divergent yet overall cooperative manner to orchestrate feeding behavior through transitions between various appetitive states, and thus offer novel insights into LHA allostatic control of appetite.
Collapse
Affiliation(s)
| | | | - Alexander W Johnson
- Department of Psychology; Neuroscience Program, Michigan State University, East Lansing.
| |
Collapse
|
14
|
Mediavilla C. Bidirectional gut-brain communication: A role for orexin-A. Neurochem Int 2020; 141:104882. [PMID: 33068686 DOI: 10.1016/j.neuint.2020.104882] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/02/2020] [Accepted: 10/11/2020] [Indexed: 02/06/2023]
Abstract
It is increasingly evident that bidirectional gut-brain signaling provides a communication pathway that uses neural, hormonal, and immunological routes to regulate homeostatic mechanisms such as hunger/satiety as well as emotions and inflammation. Hence, disruption of the gut-brain axis can cause numerous pathophysiologies, including obesity and intestinal inflammatory diseases. One chemical mediator in the gut-brain axis is orexin-A, given that hypothalamic orexin-A affects gastrointestinal motility and secretion, and peripheral orexin in the intestinal mucosa can modulate brain functions, making possible an orexinergic gut-brain network. It has been proposed that orexin-A acts on this axis to regulate nutritional processes, such as short-term intake, gastric acid secretion, and motor activity associated with the cephalic phase of feeding. Orexin-A has also been related to stress systems and stress responses via the hypothalamic-pituitary-adrenal axis. Recent studies on the relationship of orexin with immune system-brain communications in an animal model of colitis suggested an immunomodulatory role for orexin-A in signaling and responding to infection by reducing the production of pro-inflammatory cytokines (e.g., tumor necrosis factor α, interleukin-6, and monocyte chemoattractant protein-1). These studies suggested that orexin administration might be of potential therapeutic value in irritable bowel syndrome or chronic intestinal inflammatory diseases, in which gastrointestinal symptoms frequently coexist with behavioral disorders, including loss of appetite, anxiety, depression, and sleeping disorders. Interventions in the orexinergic system have been proposed as a therapeutic approach to these diseases and for the treatment of chemotherapeutic drug-related hyperalgesia and fatigue in cancer patients.
Collapse
Affiliation(s)
- Cristina Mediavilla
- Department of Psychobiology, and Mind, Brain, and Behavior Research Center (CIMCYC), University of Granada, Spain.
| |
Collapse
|
15
|
Perez-Bonilla P, Santiago-Colon K, Leinninger GM. Lateral hypothalamic area neuropeptides modulate ventral tegmental area dopamine neurons and feeding. Physiol Behav 2020; 223:112986. [PMID: 32492498 DOI: 10.1016/j.physbeh.2020.112986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 01/26/2023]
Abstract
Understanding how the brain coordinates energy status with the motivation to eat is crucial to identify strategies to improve disordered body weight. The ventral tegmental area (VTA), known as the core of the mesolimbic system, is of particular interest in this regard because it controls the motivation to consume palatable, calorie-dense foods and to engage in volitional activity. The VTA is largely composed of dopamine (DA) neurons, but modulating these DA neurons has been alternately linked with promoting and suppressing feeding, suggesting heterogeneity in their function. Subsets of VTA DA neurons have recently been described based on their anatomical distribution, electrophysiological features, connectivity and molecular expression, but to date there are no signatures to categorize how DA neurons control feeding. Assessing the neuropeptide receptors expressed by VTA DA neurons may be useful in this regard, as many neuropeptides mediate anorexic or orexigenic responses. In particular, the lateral hypothalamic area (LHA) releases a wide variety of feeding-modulating neuropeptides to the VTA. Since VTA neurons intercept LHA neuropeptides known to either evoke or suppress feeding, expression of the cognate neuropeptide receptors within the VTA may point to VTA DA neuronal mechanisms to promote or suppress feeding, respectively. Here we review the role of the VTA in energy balance and the LHA neuropeptide signaling systems that act in the VTA, whose receptors might be used to classify how VTA DA neurons contribute to energy balance.
Collapse
Affiliation(s)
- Patricia Perez-Bonilla
- Neuroscience Graduate Program, USA; Pharmacology and Toxicology Graduate Program, USA; Michigan State University, East Lansing, MI 48114, USA
| | - Krystal Santiago-Colon
- Department of Biology, University of Puerto Rico - Cayey, USA; Bridge to the PhD in Neuroscience Program, USA
| | - Gina M Leinninger
- Department of Physiology, USA; Michigan State University, East Lansing, MI 48114, USA.
| |
Collapse
|
16
|
Linehan V, Fang LZ, Parsons MP, Hirasawa M. High-fat diet induces time-dependent synaptic plasticity of the lateral hypothalamus. Mol Metab 2020; 36:100977. [PMID: 32277924 PMCID: PMC7170999 DOI: 10.1016/j.molmet.2020.100977] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/28/2020] [Accepted: 03/10/2020] [Indexed: 11/15/2022] Open
Abstract
Objective Orexin (ORX) and melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus are critical regulators of energy homeostasis and are thought to differentially contribute to diet-induced obesity. However, it is unclear whether the synaptic properties of these cells are altered by obesogenic diets over time. Methods Rats and mice were fed a control chow or palatable high-fat diet (HFD) for various durations and then synaptic properties of ORX and MCH neurons were examined using exvivo whole-cell patch clamp recording. Confocal imaging was performed to assess the number of excitatory synaptic contacts to these neurons. Results ORX neurons exhibited a transient increase in spontaneous excitatory transmission as early as 1 day up to 1 week of HFD, which returned to control levels with prolonged feeding. Conversely, HFD induced a delayed increase in excitatory synaptic transmission to MCH neurons, which progressively increased as HFD became chronic. This increase occurred before the onset of significant weight gain. These synaptic changes appeared to be due to altered postsynaptic sensitivity or the number of active synaptic contacts depending on cell type and feeding duration. However, HFD induced no change in inhibitory transmission in either cell type at any time point. Conclusions These results suggest that the effects of HFD on feeding-related neurons are cell type-specific and dynamic. This highlights the importance of considering the feeding duration for research and weight loss interventions. ORX neurons may contribute to early hyperphagia, whereas MCH neurons may play a role in the onset and long-term maintenance of diet-induced obesity. High-fat diet increases excitatory transmission to orexin and MCH neurons. Increased excitatory drive to orexin neurons occurs within the first week but is transient. Excitatory synapses to MCH neurons increase with prolonged high-fat diet. Excitatory changes in MCH neurons are delayed but precede significant weight gain. These synaptic changes may contribute to the development and the maintenance of obesity.
Collapse
Affiliation(s)
- Victoria Linehan
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada
| | - Lisa Z Fang
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada.
| |
Collapse
|
17
|
Han D, Han F, Shi Y, Zheng S, Wen L. Mechanisms of Memory Impairment Induced by Orexin-A via Orexin 1 and Orexin 2 Receptors in Post-traumatic Stress Disorder Rats. Neuroscience 2020; 432:126-136. [PMID: 32112915 DOI: 10.1016/j.neuroscience.2020.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022]
Abstract
Post-traumatic stress disorder (PTSD) patients exhibit abnormal learning and memory. Axons from orexin neurons in the lateral hypothalamus innervate the hippocampus, modulating learning and memory via the orexin 1 and 2 receptors (OX1R and OX2R). However, the role of the orexin system in the learning and memory dysfunction observed in PTSD is unknown. This was investigated in the present study using PTSD animal model-single prolonged stress (SPS) rats. Spatial learning and memory in the rats were evaluated with the Morris water maze (MWM) test; changes in body weight and food intake were recorded to assess changes in appetite; and the expression of orexin-A and its receptors in the hypothalamus and hippocampus was examined and quantified by immunohistochemistry, western blotting and real-time PCR. The results showed that spatial memory was impaired and food intake was decreased in SPS rats; this was accompanied by downregulation of orexin-A in the hypothalamus and upregulation of OX1R and OX2R in the hippocampus and of OX1R in the hypothalamus. Intracerebroventricular administration of orexin-A improved spatial memory and enhanced appetite in SPS rats and partly reversed the increases in OX1R and OX2R levels in the hippocampus and hypothalamus. These results suggest that the orexin system plays a critical role in the memory and appetite dysfunction observed in PTSD.
Collapse
Affiliation(s)
- Dan Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China; Department of Neonatology, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping Area, Shenyang, PR China
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| | - Shilei Zheng
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Lili Wen
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| |
Collapse
|
18
|
Weidner J, Jensen CH, Giske J, Eliassen S, Jørgensen C. Hormones as adaptive control systems in juvenile fish. Biol Open 2020; 9:bio046144. [PMID: 31996351 PMCID: PMC7044463 DOI: 10.1242/bio.046144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Growth is an important theme in biology. Physiologists often relate growth rates to hormonal control of essential processes. Ecologists often study growth as a function of gradients or combinations of environmental factors. Fewer studies have investigated the combined effects of environmental and hormonal control on growth. Here, we present an evolutionary optimization model of fish growth that combines internal regulation of growth by hormone levels with the external influence of food availability and predation risk. The model finds a dynamic hormone profile that optimizes fish growth and survival up to 30 cm, and we use the probability of reaching this milestone as a proxy for fitness. The complex web of interrelated hormones and other signalling molecules is simplified to three functions represented by growth hormone, thyroid hormone and orexin. By studying a range from poor to rich environments, we find that the level of food availability in the environment results in different evolutionarily optimal strategies of hormone levels. With more food available, higher levels of hormones are optimal, resulting in higher food intake, standard metabolism and growth. By using this fitness-based approach we also find a consequence of evolutionary optimization of survival on optimal hormone use. Where foraging is risky, the thyroid hormone can be used strategically to increase metabolic potential and the chance of escaping from predators. By comparing model results to empirical observations, many mechanisms can be recognized, for instance a change in pace-of-life due to resource availability, and reduced emphasis on reserves in more stable environments.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jacqueline Weidner
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| | | | - Jarl Giske
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| | - Sigrunn Eliassen
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| | - Christian Jørgensen
- University of Bergen, Department of Biological Sciences, Postboks 7803, N-5020 Bergen, Norway
| |
Collapse
|
19
|
Liu L, Wang Q, Liu A, Lan X, Huang Y, Zhao Z, Jie H, Chen J, Zhao Y. Physiological Implications of Orexins/Hypocretins on Energy Metabolism and Adipose Tissue Development. ACS OMEGA 2020; 5:547-555. [PMID: 31956801 PMCID: PMC6964296 DOI: 10.1021/acsomega.9b03106] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/27/2019] [Indexed: 05/09/2023]
Abstract
Orexins/hypocretins and their receptors (OXRs) are ubiquitously distributed throughout the nervous system and peripheral tissues. Recently, various reports have indicated that orexins play regulatory roles in numerous physiological processes involved in obesity, energy homeostasis, sleep-wake cycle, analgesia, alcoholism, learning, and memory. This review aims to outline recent progress in the research and development of orexins used in biochemical signaling pathways, secretion pathways, and the regulation of energy metabolism/adipose tissue development. Orexins regulate a variety of physiological functions in the body by activating phospholipase C/protein kinase C and AC/cAMP/PKA pathways, through receptors coupled to Gq and Gi/Gs, respectively. The secretion of orexins is modulated by blood glucose, blood lipids, hormones, and neuropeptides. Orexins have critical functions in energy metabolism, regulating both feeding behavior and energy expenditure. Increasing the sensitivity of orexin-coupled hypothalamic neurons concurrently enhances spontaneous physical activity, non-exercise activity thermogenesis, white adipose tissue lipolysis, and brown adipose tissue thermogenesis. With this comprehensive review of the current literature on the subject, we hope to provide an integrated perspective for the prevention/treatment of obesity.
Collapse
Affiliation(s)
- Lingbin Liu
- College of Animal
Science and Technology, Chongqing Key Laboratory of Forage & Herbivore,
Chongqing Engineering Research Center for Herbivores Resource Protection
and Utilization, Southwest University, Beibei, 400715 Chongqing, P. R. China
- E-mail: (L.L.)
| | - Qigui Wang
- ChongQing Academy
of Animal Sciences, Rongchang, 402460 Chongqing, P. R. China
| | - Anfang Liu
- College of Animal Science, Southwest University, Rongchang Campus, Rongchang, 402460 Chongqing, P.R. China
| | - Xi Lan
- College of Animal
Science and Technology, Chongqing Key Laboratory of Forage & Herbivore,
Chongqing Engineering Research Center for Herbivores Resource Protection
and Utilization, Southwest University, Beibei, 400715 Chongqing, P. R. China
| | - Yongfu Huang
- College of Animal
Science and Technology, Chongqing Key Laboratory of Forage & Herbivore,
Chongqing Engineering Research Center for Herbivores Resource Protection
and Utilization, Southwest University, Beibei, 400715 Chongqing, P. R. China
| | - Zhongquan Zhao
- College of Animal
Science and Technology, Chongqing Key Laboratory of Forage & Herbivore,
Chongqing Engineering Research Center for Herbivores Resource Protection
and Utilization, Southwest University, Beibei, 400715 Chongqing, P. R. China
| | - Hang Jie
- Chongqing Institute of Medicinal Plant
Cultivation, Nanchuan, 408435 Chongqing, P.R. China
| | - Juncai Chen
- College of Animal
Science and Technology, Chongqing Key Laboratory of Forage & Herbivore,
Chongqing Engineering Research Center for Herbivores Resource Protection
and Utilization, Southwest University, Beibei, 400715 Chongqing, P. R. China
| | - Yongju Zhao
- College of Animal
Science and Technology, Chongqing Key Laboratory of Forage & Herbivore,
Chongqing Engineering Research Center for Herbivores Resource Protection
and Utilization, Southwest University, Beibei, 400715 Chongqing, P. R. China
- E-mail: (Y.Z.)
| |
Collapse
|
20
|
Straat ME, Schinkelshoek MS, Fronczek R, Lammers GJ, Rensen PCN, Boon MR. Role of Brown Adipose Tissue in Adiposity Associated With Narcolepsy Type 1. Front Endocrinol (Lausanne) 2020; 11:145. [PMID: 32373062 PMCID: PMC7176868 DOI: 10.3389/fendo.2020.00145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/02/2020] [Indexed: 11/23/2022] Open
Abstract
Narcolepsy type 1 is a neurological sleep-wake disorder caused by the destruction of orexin (hypocretin)-producing neurons. These neurons are particularly located in the lateral hypothalamus and have widespread projections throughout the brain, where they are involved, e.g., in the regulation of the sleep-wake cycle and appetite. Interestingly, a higher prevalence of obesity has been reported in patients with narcolepsy type 1 compared to healthy controls, despite a normal to decreased food intake and comparable physical activity. This suggests the involvement of tissues implicated in total energy expenditure, including skeletal muscle, liver, white adipose tissue (WAT), and brown adipose tissue (BAT). Recent evidence from pre-clinical studies with orexin knock-out mice demonstrates a crucial role for the orexin system in the functionality of brown adipose tissue (BAT), probably through multiple pathways. Since BAT is a highly metabolically active organ that combusts fatty acids and glucose toward heat, thereby contributing to energy metabolism, this raises the question of whether BAT plays a role in the development of obesity and related metabolic diseases in narcolepsy type 1. BAT is densely innervated by the sympathetic nervous system that activates BAT, for instance, following cold exposure. The sympathetic outflow toward BAT is mainly mediated by the dorsomedial, ventromedial, arcuate, and paraventricular nuclei in the hypothalamus. This review focuses on the current knowledge on the role of the orexin system in the control of energy balance, with specific focus on BAT metabolism and adiposity in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Maaike E. Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Maaike E. Straat
| | - Mink S. Schinkelshoek
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
- Sleep Wake Centre SEIN, Heemstede, Netherlands
| | - Rolf Fronczek
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
- Sleep Wake Centre SEIN, Heemstede, Netherlands
| | - Gerrit Jan Lammers
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
- Sleep Wake Centre SEIN, Heemstede, Netherlands
| | - Patrick C. N. Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
21
|
Milbank E, López M. Orexins/Hypocretins: Key Regulators of Energy Homeostasis. Front Endocrinol (Lausanne) 2019; 10:830. [PMID: 31920958 PMCID: PMC6918865 DOI: 10.3389/fendo.2019.00830] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 11/13/2019] [Indexed: 12/29/2022] Open
Abstract
Originally described to be involved in feeding regulation, orexins/hypocretins are now also considered as major regulatory actors of numerous biological processes, such as pain, sleep, cardiovascular function, neuroendocrine regulation, and energy expenditure. Therefore, they constitute one of the most pleiotropic families of hypothalamic neuropeptides. Although their orexigenic effect is well documented, orexins/hypocretins also exert central effects on energy expenditure, notably on the brown adipose tissue (BAT) thermogenesis. A better comprehension of the underlying mechanisms and potential interactions with other hypothalamic molecular pathways involved in the modulation of food intake and thermogenesis, such as AMP-activated protein kinase (AMPK) and endoplasmic reticulum (ER) stress, is essential to determine the exact implication and pathophysiological relevance of orexins/hypocretins on the control of energy balance. Here, we will review the actions of orexins on energy balance, with special focus on feeding and brown fat function.
Collapse
Affiliation(s)
- Edward Milbank
- Department of Physiology, CIMUS, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- *Correspondence: Edward Milbank
| | - Miguel López
- Department of Physiology, CIMUS, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Miguel López
| |
Collapse
|
22
|
Activation of orexin-1 receptors in the amygdala enhances feeding in the diet-induced obesity rats: Blockade with μ-opioid antagonist. Biochem Biophys Res Commun 2018; 503:3186-3191. [DOI: 10.1016/j.bbrc.2018.08.120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 08/18/2018] [Indexed: 12/31/2022]
|
23
|
Barbier M, Fellmann D, Risold PY. Morphofunctional Organization of the Connections From the Medial and Intermediate Parts of the Central Nucleus of the Amygdala Into Distinct Divisions of the Lateral Hypothalamic Area in the Rat. Front Neurol 2018; 9:688. [PMID: 30210427 PMCID: PMC6119805 DOI: 10.3389/fneur.2018.00688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/30/2018] [Indexed: 11/23/2022] Open
Abstract
Projections from the central nucleus of the amygdala (CEA) into the lateral hypothalamic area (LHA) show a very complex pattern. After injection of an anterograde tracer (Phaseolus vulgaris leucoagglutinin—PHAL) into the medial and intermediate parts of the CEA, we observed that labeled axons converged onto the caudal lateral LHA but provided distinct patterns in rostral tuberal regions. These projections were compared to that of neurons containing the peptides “melanin-concentrating hormone” (MCH) or hypocretin (Hcrt). Because the distribution of these neurons is stereotyped, it was possible to characterize distinct divisions into the LHA. Some of them in the rostral tuberal LHA [the dorsal (LHAd) and suprafornical regions (LHAs)] received a distinct innervation by projections that originated from neurons in respectively anterior or posterior regions of the medial part (CEAm) or from the intermediate part (CEAi) of the central nucleus. Therefore, this work illustrates that projections from the CEAm and CEAi converge into the caudal lateral LHA but diverge into the rostral tuberal LHA.
Collapse
Affiliation(s)
- Marie Barbier
- Laboratoire de Neurosciences Intégratives et Cliniques, EA481, UFR Sciences Médicales et Pharmaceutiques, Université de Bourgogne Franche-Comté, Besançon, France
| | - Dominique Fellmann
- Laboratoire de Neurosciences Intégratives et Cliniques, EA481, UFR Sciences Médicales et Pharmaceutiques, Université de Bourgogne Franche-Comté, Besançon, France
| | - Pierre-Yves Risold
- Laboratoire de Neurosciences Intégratives et Cliniques, EA481, UFR Sciences Médicales et Pharmaceutiques, Université de Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
24
|
Risco S, Mediavilla C. Orexin A in the ventral tegmental area enhances saccharin-induced conditioned flavor preference: The role of D1 receptors in central nucleus of amygdala. Behav Brain Res 2018; 348:192-200. [DOI: 10.1016/j.bbr.2018.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/24/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022]
|
25
|
The weaning period promotes alterations in the orexin neuronal population of rats in a suckling-dependent manner. Brain Struct Funct 2018; 223:3739-3755. [DOI: 10.1007/s00429-018-1723-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 07/07/2018] [Indexed: 10/28/2022]
|
26
|
Ericson MD, Haskell-Luevano C. A Review of Single-Nucleotide Polymorphisms in Orexigenic Neuropeptides Targeting G Protein-Coupled Receptors. ACS Chem Neurosci 2018; 9:1235-1246. [PMID: 29714060 DOI: 10.1021/acschemneuro.8b00151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many physiological pathways are involved in appetite, food intake, and the maintenance of energy homeostasis. In particular, neuropeptides within the central nervous system have been demonstrated to be critical signaling molecules for modulating appetite. Both anorexigenic (appetite-decreasing) and orexigenic (appetite-stimulating) neuropeptides have been described. The biological effects of these neuropeptides can be observed following central administration in animal models. This review focuses on single nucleotide polymorphisms (SNPs) in six orexigenic neuropeptides: agouti-related protein (AGRP), galanin, melanin concentrating hormone (MCH), neuropeptide Y (NPY), orexin A, and orexin B. Following a brief summary of the neuropeptides and their orexigenic activities, reports associating SNPs within the orexigenic neuropeptides to energy homeostasis, food intake, obesity, and BMI in humans are reviewed. Additionally, the NIH tool Variation Viewer was utilized to identify missense SNPs within the mature, biologically active neuropeptide sequences. For SNPs found through Variation Viewer, a concise discussion on relevant pharmacological structure-activity relationship studies for select SNPs is included. This review is meant to update reported orexigenic neuropeptide SNPs and demonstrate the potential utility of genomic sequence databases for finding SNPs that may result in altered receptor signaling for neuropeptide pathways associated with appetite.
Collapse
Affiliation(s)
- Mark D. Ericson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
27
|
Tsai MC, Huang TL. Orexin A in men with heroin use disorder undergoing methadone maintenance treatment. Psychiatry Res 2018; 264:412-415. [PMID: 29680730 DOI: 10.1016/j.psychres.2018.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 04/02/2018] [Accepted: 04/02/2018] [Indexed: 12/16/2022]
Abstract
Orexins have played a role in reward-seeking and addiction-related behavior. There are few reports in the literature on serum levels of orexins in patients with heroin use disorder (HUD) undergoing methadone maintenance treatment (MMT). The aim of this study was to investigate the serum levels of orexin A in HUD patients undergoing MMT. Fifty male HUD patients undergoing MMT and 25 healthy males were enrolled for this study. Serum orexin A were measured with assay kits. Using analysis of covariance (ANCOVA) with body mass index (BMI) adjustments, the serum levels of orexin A in HUD men undergoing MMT were found to be significantly higher than in healthy controls. In conclusion, our results suggest that MMT might increase orexin A levels in HUD patients.
Collapse
Affiliation(s)
- Meng-Chang Tsai
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Tiao-Lai Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC.
| |
Collapse
|
28
|
Abstract
The hypothalamus is involved in the regulation of homeostatic mechanisms and migraine-related trigeminal nociception and as such has been hypothesized to play a central role in the migraine syndrome from the earliest stages of the attack. The hypothalamus hosts many key neuropeptide systems that have been postulated to play a role in this pathophysiology. Such neuropeptides include but are not exclusive too orexins, oxytocin, neuropeptide Y, and pituitary adenylate cyclase activating protein, which will be the focus of this review. Each of these peptides has its own unique physiological role and as such many preclinical studies have been conducted targeting these peptide systems with evidence supporting their role in migraine pathophysiology. Preclinical studies have also begun to explore potential therapeutic compounds targeting these systems with some success in all cases. Clinical efficacy of dual orexin receptor antagonists and intranasal oxytocin have been tested; however, both have yet to demonstrate clinical effect. Despite this, there were limitations in these cases and strong arguments can be made for the further development of intranasal oxytocin for migraine prophylaxis. Regarding neuropeptide Y, work has yet to begun in a clinical setting, and clinical trials for pituitary adenylate cyclase activating protein are just beginning to be established with much optimism. Regardless, it is becoming increasingly clear the prominent role that the hypothalamus and its peptide systems have in migraine pathophysiology. Much work is required to better understand this system and the early stages of the attack to develop more targeted and effective therapies aimed at reducing attack susceptibility with the potential to prevent the attack all together.
Collapse
Affiliation(s)
- Lauren C Strother
- Headache Group, Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Anan Srikiatkhachorn
- International Medical College, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Weera Supronsinchai
- Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Pathumwan, Bangkok, Thailand.
| |
Collapse
|
29
|
Mäkelä KA, Karhu T, Jurado Acosta A, Vakkuri O, Leppäluoto J, Herzig KH. Plasma Orexin-A Levels Do Not Undergo Circadian Rhythm in Young Healthy Male Subjects. Front Endocrinol (Lausanne) 2018; 9:710. [PMID: 30568633 PMCID: PMC6289979 DOI: 10.3389/fendo.2018.00710] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/12/2018] [Indexed: 11/22/2022] Open
Abstract
Orexin-A (OXA) has been originally isolated from a precursor peptide prepro-orexin from the lateral hypothalamus. The orexin system has been attributed to important functions in sleep, arousal and regulation of energy homeostasis. In addition to its high levels in cerebrospinal fluid, OXA is present in blood. However, reported peptide concentrations in plasma vary significantly depending on the method used. Therefore, a specific and sensitive OXA radioimmunoassay (RIA) with solid phase extraction method was developed to determine whether plasma OXA concentrations is affected by acute feeding and/or wake and sleep in young healthy males. Blood samples were collected for 24 h from nine healthy males (aged 20-24 years; BMI 20.7-26.5) every 2 h starting at 11 a.m. Food was served at 12 p.m, 5:30 p.m, 8 p.m and 8 a.m and the sleep time was between 10 p.m and 7 a.m. Plasma samples were analyzed in addition for cortisol and melatonin levels. Blood pressure was monitored through the experimental period. OXA antibody was raised in rabbits. OXA antiserum had only minor cross-reactivity with prepro-orexin precursor (<0.001%), amino-terminal peptide (<0.001%), carboxy-terminal peptide (0.001%), and orexin-B (0.3%) with high sensitivity (0.15 pg/tube). Plasma OXA levels varied between 0.5 and 16 pg/ml in seven subjects and were undetectable (below 0.5 pg/ml) in two subjects. The OXA concentrations did not correlate to feeding nor wake/sleep, whereas cortisol, melatonin and mean arterial blood pressure presented a clear circadian rhythm in each subject. In conclusion, OXA is present in blood in low amounts and its levels do not follow autonomic nor neuroendocrine circadian rhythms. Thereby, studies examining regulatory mechanisms and influences of OXA from blood samples should interpret results very cautiously.
Collapse
Affiliation(s)
- Kari A. Mäkelä
- Research Unit of Biomedicine, Physiology, University of Oulu, Oulu, Finland
- *Correspondence: Karl-Heinz Herzig
| | - Toni Karhu
- Research Unit of Biomedicine, Physiology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Olavi Vakkuri
- Research Unit of Biomedicine, Physiology, University of Oulu, Oulu, Finland
| | - Juhani Leppäluoto
- Research Unit of Biomedicine, Physiology, University of Oulu, Oulu, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Physiology, University of Oulu, Oulu, Finland
- Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
- Kari A. Mäkelä
| |
Collapse
|
30
|
Affiliation(s)
- I O Ebrahim
- Department of Psychiatry, Lane Fox Unit, St Thomas' Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | | | | | | | | |
Collapse
|
31
|
Baimel C, Borgland SL. Hypocretin/Orexin and Plastic Adaptations Associated with Drug Abuse. Curr Top Behav Neurosci 2017; 33:283-304. [PMID: 28303403 DOI: 10.1007/7854_2016_44] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are a critical part of the neural circuits that underlie reward learning and motivation. Dopamine neurons send dense projections throughout the brain and recent observations suggest that both the intrinsic properties and the functional output of dopamine neurons are dependent on projection target and are subject to neuromodulatory influences. Lateral hypothalamic hypocretin (also termed orexin) neurons project to the VTA and contain both hypocretin and dynorphin peptides in the same dense core vesicles suggesting they may be co-released. Hypocretin peptides act at excitatory Gαq protein-coupled receptors and dynorphin acts at inhibitory Gαi/o protein-coupled receptors, which are both expressed on subpopulations of dopamine neurons. This review describes a role for neuromodulation of dopamine neurons and the influence on motivated behaviour in response to natural and drug rewards.
Collapse
Affiliation(s)
- Corey Baimel
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1
| | - Stephanie L Borgland
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1.
| |
Collapse
|
32
|
Lau BK, Cota D, Cristino L, Borgland SL. Endocannabinoid modulation of homeostatic and non-homeostatic feeding circuits. Neuropharmacology 2017; 124:38-51. [PMID: 28579186 DOI: 10.1016/j.neuropharm.2017.05.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 02/03/2023]
Abstract
The endocannabinoid system has emerged as a key player in the control of eating. Endocannabinoids, including 2-arachidonoylglycerol (2-AG) and anandamide (AEA), modulate neuronal activity via cannabinoid 1 receptors (CB1Rs) in multiple nuclei of the hypothalamus to induce or inhibit food intake depending on nutritional and hormonal status, suggesting that endocannabinoids may act in the hypothalamus to integrate different types of signals informing about the animal's energy needs. In the mesocorticolimbic system, (endo)cannabinoids modulate synaptic transmission to promote dopamine release in response to palatable food. In addition, (endo)cannabinoids act within the nucleus accumbens to increase food's hedonic impact; although this effect depends on activation of CB1Rs at excitatory, but not inhibitory inputs in the nucleus accumbens. While hyperactivation of the endocannabinoid system is typically associated with overeating and obesity, much evidence has emerged in recent years suggesting a more complicated system than first thought - endocannabinoids promote or suppress feeding depending on cell and input type, or modulation by various neuronal or hormonal signals. This review presents our latest knowledge of the endocannabinoid system in non-homeostatic and homeostatic feeding circuits. In particular, we discuss the functional role and cellular mechanism of action by endocannabinoids within the hypothalamus and mesocorticolimbic system, and how these are modulated by neuropeptide signals related to feeding. In light of recent advances and complexity in the field, we review cannabinoid-based therapeutic strategies for the treatment of obesity and how peripheral restriction of CB1R antagonists may provide a different mechanism of weight loss without the central adverse effects. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Benjamin K Lau
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Daniela Cota
- INSERM U1215, Université de Bordeaux, NeuroCentre Magendie, 146, rue Léo Saignat, 33077 Bordeaux, France
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry of CNR, Viale Campi Flegrei, 34, 80078 Pozzuoli, Napoli, Italy
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
33
|
Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, Akerman S. Pathophysiology of Migraine: A Disorder of Sensory Processing. Physiol Rev 2017; 97:553-622. [PMID: 28179394 PMCID: PMC5539409 DOI: 10.1152/physrev.00034.2015] [Citation(s) in RCA: 1036] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Plaguing humans for more than two millennia, manifest on every continent studied, and with more than one billion patients having an attack in any year, migraine stands as the sixth most common cause of disability on the planet. The pathophysiology of migraine has emerged from a historical consideration of the "humors" through mid-20th century distraction of the now defunct Vascular Theory to a clear place as a neurological disorder. It could be said there are three questions: why, how, and when? Why: migraine is largely accepted to be an inherited tendency for the brain to lose control of its inputs. How: the now classical trigeminal durovascular afferent pathway has been explored in laboratory and clinic; interrogated with immunohistochemistry to functional brain imaging to offer a roadmap of the attack. When: migraine attacks emerge due to a disorder of brain sensory processing that itself likely cycles, influenced by genetics and the environment. In the first, premonitory, phase that precedes headache, brain stem and diencephalic systems modulating afferent signals, light-photophobia or sound-phonophobia, begin to dysfunction and eventually to evolve to the pain phase and with time the resolution or postdromal phase. Understanding the biology of migraine through careful bench-based research has led to major classes of therapeutics being identified: triptans, serotonin 5-HT1B/1D receptor agonists; gepants, calcitonin gene-related peptide (CGRP) receptor antagonists; ditans, 5-HT1F receptor agonists, CGRP mechanisms monoclonal antibodies; and glurants, mGlu5 modulators; with the promise of more to come. Investment in understanding migraine has been very successful and leaves us at a new dawn, able to transform its impact on a global scale, as well as understand fundamental aspects of human biology.
Collapse
Affiliation(s)
- Peter J Goadsby
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Philip R Holland
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Margarida Martins-Oliveira
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Jan Hoffmann
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Christoph Schankin
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Simon Akerman
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
34
|
Mayannavar S, Rashmi KS, Rao YD, Yadav S, Ganaraja B. Effect of Orexin A antagonist (SB-334867) infusion into the nucleus accumbens on consummatory behavior and alcohol preference in Wistar rats. Indian J Pharmacol 2017; 48:53-8. [PMID: 26997723 PMCID: PMC4778208 DOI: 10.4103/0253-7613.174528] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Objective: Nucleus accumbens (NAcc) has a role in addiction and ingestive behavior. In order to assess orexinergic system involved in this, we infused Orexin A antagonist and assessed the effect on food intake fluid intake and alcohol preference in Wistar rats. Materials and Methods: Inbred Wistar rats (n = 54) were divided into control and experimental groups (low dose and high dose). Using stereotaxic method, guide cannula was set in place bilaterally to reach NAcc. Low dose (3 ng) and high dose (6 ng) of Orexin A antagonist (SB-334867) was infused, and the food consumption, water intake and alcohol intake, and two bottle free choice preference test for alcohol were carried out in the experimental group. The control group received saline infusion and rest of the methods followed were same. The measurements were carried out immediately after the infusion, at 1 h, 2 h, 4 h, and for the whole day and represented in the figure and tables. Results: A decrease in water intake observed immediately after the infusion in 1st h (P < 0.05) and 2nd h (P < 0.01), which was more in high dose group compared to low dose and controls. Alcohol intake was also following the same pattern. In two bottle free choice, rats did not show any specific preference to alcohol. Conclusion: There was dose dependent reduction in intake of food and fluids in treated rats. This suggested a possible role for orexinergic system in ingestive behavior. However, Orexin A may not have a role in modulation of alcohol addiction by the rewarding center NAcc.
Collapse
Affiliation(s)
- Santosh Mayannavar
- Department of Physiology, Kasturba Medical College (A Unit of Manipal University), Mangalore, India
| | - K S Rashmi
- Department of Physiology, Kasturba Medical College (A Unit of Manipal University), Mangalore, India
| | - Yalla Durga Rao
- Department of Biochemistry, Kasturba Medical College (A Unit of Manipal University), Mangalore, Karnataka, India
| | - Saraswati Yadav
- Department of Physiology, Kasturba Medical College (A Unit of Manipal University), Mangalore, India
| | - B Ganaraja
- Department of Physiology, Kasturba Medical College (A Unit of Manipal University), Mangalore, India
| |
Collapse
|
35
|
Ueta Y, Ozaki Y, Saito J, Onaka T. Involvement of Novel Feeding-Related Peptides in Neuroendocrine Response to Stress. Exp Biol Med (Maywood) 2016; 228:1168-74. [PMID: 14610256 DOI: 10.1177/153537020322801011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Various stressors are known to cause eating disorders. However, it is not known in detail about the neural network and molecular mechanism that are involved in the stress-induced changes of feeding behavior in the central nervous system. Many novel feeding-regulated peptides such as orexins/hypocretins and ghrelin have been discovered since the discovery of leptin derived from adipocytes as a product of the ob gene. These novel peptides were identified as endogenous ligands of orphan G protein-coupled receptors. The accumulating evidence reveals that these peptides may be involved in stress responses via the central nervous system, as well as feeding behavior. The possible involvement of novel feeding-related peptides in neuroendocrine responses to stress is reviewed here.
Collapse
Affiliation(s)
- Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan.
| | | | | | | |
Collapse
|
36
|
|
37
|
Milagro FI, Martínez JA. Orexin A and B are Involved in the Regulation of Body Temperature and Glucose Homeostasis in Rats. Nutr Neurosci 2016. [DOI: 10.1080/1028415x.2000.11747344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
38
|
Tang Y, Benusiglio D, Grinevich V, Lin L. Distinct Types of Feeding Related Neurons in Mouse Hypothalamus. Front Behav Neurosci 2016; 10:91. [PMID: 27242460 PMCID: PMC4870269 DOI: 10.3389/fnbeh.2016.00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022] Open
Abstract
The last two decades of research provided evidence for a substantial heterogeneity among feeding-related neurons (FRNs) in the hypothalamus. However, it remains unclear how FRNs differ in their firing patterns during food intake. Here, we investigated the relationship between the activity of neurons in mouse hypothalamus and their feeding behavior. Using tetrode-based in vivo recording technique, we identified various firing patterns of hypothalamic FRNs, which, after the initiation of food intake, can be sorted into four types: sharp increase (type I), slow increase (type II), sharp decrease (type III), and sustained decrease (type IV) of firing rates. The feeding-related firing response of FRNs was rigidly related to the duration of food intake and, to a less extent, associated with the type of food. The majority of these FRNs responded to glucose and leptin and exhibited electrophysiological characteristics of putative GABAergic neurons. In conclusion, our study demonstrated the diversity of neurons in the complex hypothalamic network coordinating food intake.
Collapse
Affiliation(s)
- Yan Tang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Science and the Collaborative Innovation Center for Brain Science, Institute of Brain Functional Genomics, East China Normal UniversityShanghai, China; Schaller Research Group on Neuropeptides at German Cancer Research Center, Central Institute of Mental Health, and Cell Networks Cluster of Excellence at the University of HeidelbergHeidelberg, Germany
| | - Diego Benusiglio
- Schaller Research Group on Neuropeptides at German Cancer Research Center, Central Institute of Mental Health, and Cell Networks Cluster of Excellence at the University of Heidelberg Heidelberg, Germany
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides at German Cancer Research Center, Central Institute of Mental Health, and Cell Networks Cluster of Excellence at the University of Heidelberg Heidelberg, Germany
| | - Longnian Lin
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Science and the Collaborative Innovation Center for Brain Science, Institute of Brain Functional Genomics, East China Normal University Shanghai, China
| |
Collapse
|
39
|
Hunt NJ, Russell B, Du MK, Waters KA, Machaalani R. Changes in orexinergic immunoreactivity of the piglet hypothalamus and pons after exposure to chronic postnatal nicotine and intermittent hypercapnic hypoxia. Eur J Neurosci 2016; 43:1612-22. [DOI: 10.1111/ejn.13246] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/10/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Nicholas J. Hunt
- Department of Medicine; Central Clinical School; University of Sydney; Camperdown NSW Australia
- BOSCH Institute of Biomedical Research; University of Sydney; Camperdown NSW Australia
| | - Benjamin Russell
- Summer Research Scholarship Program; Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Man K. Du
- Department of Pathology; Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Karen A. Waters
- Department of Medicine; Central Clinical School; University of Sydney; Camperdown NSW Australia
- BOSCH Institute of Biomedical Research; University of Sydney; Camperdown NSW Australia
- The Children's Hospital; Westmead NSW Australia
| | - Rita Machaalani
- Department of Medicine; Central Clinical School; University of Sydney; Camperdown NSW Australia
- BOSCH Institute of Biomedical Research; University of Sydney; Camperdown NSW Australia
- The Children's Hospital; Westmead NSW Australia
- Room 206; SIDS and Sleep Apnoea Laboratory; University of Sydney; Blackburn Building, D06 Camperdown NSW 2006 Australia
| |
Collapse
|
40
|
Sun S, Xu L, Sun X, Guo F, Gong Y, Gao S. Orexin-A affects gastric distention sensitive neurons in the hippocampus and gastric motility and regulation by the perifornical area in rats. Neurosci Res 2016; 110:59-67. [PMID: 27080329 DOI: 10.1016/j.neures.2016.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/25/2016] [Accepted: 04/01/2016] [Indexed: 12/20/2022]
Abstract
Orexin-A is mainly produced in the lateral hypothalamus (LHA) and the perifornical area (PeF). Here, we aim to elucidate the effects of orexin-A in the hippocampus (Hi) on gastric distention (GD)-sensitive neurons and gastric motility, and potential regulation mechanisms by the PeF. Retrograde tracing and fluorescent-immunohistochemical staining were used to determine orexin-A neuronal projections. Single unit discharges in the Hi were recorded extracellularly and gastric motility in conscious rats was monitored during administration of orexin-A to the Hi or electrical stimulation of the PeF. Orexin-A administration to the Hi excited most of the GD-excitatory (GD-E) neurons and GD-inhibitory (GD-I) neurons, and increased gastric motility in a dose-dependent manner. All of effects induced by orexin-A could be partly blocked by pretreatment with orexin-A antagonist, SB-334867. Electrical stimulation of the PeF excited the majority of the orexin-A-responsive GD neurons in the Hi and promoted gastric motility. Additionally, pretreatment with SB-334867 in the Hi increased the firing rate of GDI and GDE neurons following electrical stimulation of the PeF. These findings suggest that orexin-A could regulate activities of GD-sensitive neurons and gastric motility. Furthermore, the PeF may be involved in this regulatory pathway.
Collapse
Affiliation(s)
- Shu Sun
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| | - Luo Xu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Xiangrong Sun
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| | - Feifei Guo
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Shengli Gao
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| |
Collapse
|
41
|
Interacting Neural Processes of Feeding, Hyperactivity, Stress, Reward, and the Utility of the Activity-Based Anorexia Model of Anorexia Nervosa. Harv Rev Psychiatry 2016; 24:416-436. [PMID: 27824637 PMCID: PMC5485261 DOI: 10.1097/hrp.0000000000000111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anorexia nervosa (AN) is a psychiatric illness with minimal effective treatments and a very high rate of mortality. Understanding the neurobiological underpinnings of the disease is imperative for improving outcomes and can be aided by the study of animal models. The activity-based anorexia rodent model (ABA) is the current best parallel for the study of AN. This review describes the basic neurobiology of feeding and hyperactivity seen in both ABA and AN, and compiles the research on the role that stress-response and reward pathways play in modulating the homeostatic drive to eat and to expend energy, which become dysfunctional in ABA and AN.
Collapse
|
42
|
Abstract
Sleep and energy balance are essential for health. The two processes act in concert to regulate central and peripheral homeostasis. During sleep, energy is conserved due to suspended activity, movement, and sensory responses, and is redirected to restore and replenish proteins and their assemblies into cellular structures. During wakefulness, various energy-demanding activities lead to hunger. Thus, hunger promotes arousal, and subsequent feeding, followed by satiety that promotes sleep via changes in neuroendocrine or neuropeptide signals. These signals overlap with circuits of sleep-wakefulness, feeding, and energy expenditure. Here, we will briefly review the literature that describes the interplay between the circadian system, sleep-wake, and feeding-fasting cycles that are needed to maintain energy balance and a healthy metabolic profile. In doing so, we describe the neuroendocrine, hormonal/peptide signals that integrate sleep and feeding behavior with energy metabolism.
Collapse
Affiliation(s)
- Charu Shukla
- Department of Psychiatry, VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| | - Radhika Basheer
- Department of Psychiatry, VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| |
Collapse
|
43
|
Khandekar N, Berning BA, Sainsbury A, Lin S. The role of pancreatic polypeptide in the regulation of energy homeostasis. Mol Cell Endocrinol 2015; 418 Pt 1:33-41. [PMID: 26123585 DOI: 10.1016/j.mce.2015.06.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/16/2015] [Accepted: 06/03/2015] [Indexed: 12/13/2022]
Abstract
Imbalances in normal regulation of food intake can cause obesity and related disorders. Inadequate therapies for such disorders necessitate better understanding of mechanisms that regulate energy homeostasis. Pancreatic polypeptide (PP), a robust anorexigenic hormone, effectively modulates food intake and energy homeostasis, thus potentially aiding anti-obesity therapeutics. Intra-gastric and intra-intestinal infusion of nutrients stimulate PP secretion from the gastrointestinal tract, leading to vagal stimulation that mediates complex actions via the neuropeptide Y4 receptor in arcuate nucleus of the hypothalamus, subsequently activating key hypothalamic nuclei and dorsal vagal complex of the brainstem to influence energy homeostasis and body composition. Novel studies indicate affinity of PP for the relatively underexplored neuropeptide y6 receptor, mediating actions via the suprachiasmatic nucleus and pathways involving vasoactive intestinal polypeptide and insulin like growth factor 1. This review highlights detailed mechanisms by which PP mediates its actions on energy balance through various areas in the brain.
Collapse
Affiliation(s)
- Neeta Khandekar
- Neurological Diseases Division, Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Britt A Berning
- Neurological Diseases Division, Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Amanda Sainsbury
- The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, NSW 2006, Australia
| | - Shu Lin
- Neurological Diseases Division, Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
44
|
Abstract
Initially implicated in the regulation of feeding, orexins/hypocretins are now acknowledged to play a major role in the control of a wide variety of biological processes, such as sleep, energy expenditure, pain, cardiovascular function and neuroendocrine regulation, a feature that makes them one of the most pleiotropic families of hypothalamic neuropeptides. While the orexigenic effect of orexins is well described, their central effects on energy expenditure and particularly on brown adipose tissue (BAT) thermogenesis are not totally unraveled. Better understanding of these actions and their possible interrelationship with other hypothalamic systems controlling thermogenesis, such as AMP-activated protein kinase (AMPK) and endoplasmic reticulum (ER) stress, will help to clarify the exact role and pathophysiological relevance of these neuropeptides have on energy balance.
Collapse
Affiliation(s)
- Johan Fernø
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Department of Clinical Science, K. G. Jebsen Center for Diabetes Research, University of Bergen, N-5021 Bergen, Norway.
| | - Rosa Señarís
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, 14004 Córdoba, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, Spain.
| |
Collapse
|
45
|
Sarihi A, Emam AH, Panah MH, Komaki A, Seif S, Vafaeirad M, Alaii E. Effects of activation and blockade of orexin A receptors in the medial preoptic area on food intake in male rats. Neurosci Lett 2015; 604:157-60. [DOI: 10.1016/j.neulet.2015.07.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 06/16/2015] [Accepted: 07/31/2015] [Indexed: 01/20/2023]
|
46
|
Brown JA, Woodworth HL, Leinninger GM. To ingest or rest? Specialized roles of lateral hypothalamic area neurons in coordinating energy balance. Front Syst Neurosci 2015; 9:9. [PMID: 25741247 PMCID: PMC4332303 DOI: 10.3389/fnsys.2015.00009] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/15/2015] [Indexed: 12/26/2022] Open
Abstract
Survival depends on an organism’s ability to sense nutrient status and accordingly regulate intake and energy expenditure behaviors. Uncoupling of energy sensing and behavior, however, underlies energy balance disorders such as anorexia or obesity. The hypothalamus regulates energy balance, and in particular the lateral hypothalamic area (LHA) is poised to coordinate peripheral cues of energy status and behaviors that impact weight, such as drinking, locomotor behavior, arousal/sleep and autonomic output. There are several populations of LHA neurons that are defined by their neuropeptide content and contribute to energy balance. LHA neurons that express the neuropeptides melanin-concentrating hormone (MCH) or orexins/hypocretins (OX) are best characterized and these neurons play important roles in regulating ingestion, arousal, locomotor behavior and autonomic function via distinct neuronal circuits. Recently, another population of LHA neurons containing the neuropeptide Neurotensin (Nts) has been implicated in coordinating anorectic stimuli and behavior to regulate hydration and energy balance. Understanding the specific roles of MCH, OX and Nts neurons in harmonizing energy sensing and behavior thus has the potential to inform pharmacological strategies to modify behaviors and treat energy balance disorders.
Collapse
Affiliation(s)
- Juliette A Brown
- Department of Pharmacology and Toxicology, Michigan State University East Lansing, MI, USA ; Center for Integrative Toxicology East Lansing, MI, USA
| | | | - Gina M Leinninger
- Center for Integrative Toxicology East Lansing, MI, USA ; Department of Physiology, Michigan State University East Lansing, MI, USA
| |
Collapse
|
47
|
Cho GJ, Hong HR, Kim SW, Hong SC, Oh MJ, Kim HJ. Decreased umbilical orexin-A level is associated with idiopathic polyhydramnios. Acta Obstet Gynecol Scand 2015; 94:295-300. [DOI: 10.1111/aogs.12559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 12/08/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Geum Joon Cho
- Department of Obstetrics and Gynecology; College of Medicine; Korea University; Seoul Korea
| | - Hye-Ri Hong
- Department of Obstetrics and Gynecology; School of Medicine; Kyung Hee University; Seoul Korea
| | - Suhng Wook Kim
- Department of Biomedical Science; College of Health Sciences; Korea University; Seoul Korea
| | - Soon Cheol Hong
- Department of Obstetrics and Gynecology; College of Medicine; Korea University; Seoul Korea
| | - Min-Jeong Oh
- Department of Obstetrics and Gynecology; College of Medicine; Korea University; Seoul Korea
| | - Hai Joon Kim
- Department of Obstetrics and Gynecology; College of Medicine; Korea University; Seoul Korea
| |
Collapse
|
48
|
Hunt NJ, Rodriguez ML, Waters KA, Machaalani R. Changes in orexin (hypocretin) neuronal expression with normal aging in the human hypothalamus. Neurobiol Aging 2015; 36:292-300. [DOI: 10.1016/j.neurobiolaging.2014.08.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 07/30/2014] [Accepted: 08/04/2014] [Indexed: 11/29/2022]
|
49
|
Baimel C, Bartlett SE, Chiou LC, Lawrence AJ, Muschamp JW, Patkar O, Tung LW, Borgland SL. Orexin/hypocretin role in reward: implications for opioid and other addictions. Br J Pharmacol 2015; 172:334-48. [PMID: 24641197 PMCID: PMC4292951 DOI: 10.1111/bph.12639] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/24/2014] [Accepted: 01/31/2014] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Addiction is a devastating disorder that affects 15.3 million people worldwide. While prevalent, few effective treatments exist. Orexin receptors have been proposed as a potential target for anti-craving medications. Orexins, also known as hypocretins, are neuropeptides produced in neurons of the lateral and dorsomedial hypothalamus and perifornical area, which project widely throughout the brain. The absence of orexins in rodents and humans leads to narcolepsy. However, orexins also have an established role in reward seeking. This review will discuss some of the original studies describing the roles of the orexins in reward seeking as well as specific works that were presented at the 2013 International Narcotics Research Conference. Orexin signalling can promote drug-induced plasticity of glutamatergic synapses onto dopamine neurons of the ventral tegmental area (VTA), a brain region implicated in motivated behaviour. Additional evidence suggests that orexin signalling can also promote drug seeking by initiating an endocannabinoid-mediated synaptic depression of GABAergic inputs to the VTA, and thereby disinhibiting dopaminergic neurons. Orexin neurons co-express the inhibitory opioid peptide dynorphin. It has been proposed that orexin in the VTA may not mediate reward per se, but rather occludes the 'anti-reward' effects of dynorphin. Finally, orexin signalling in the prefrontal cortex and the central amygdala is implicated in reinstatement of reward seeking. This review will highlight recent work describing the role of orexin signalling in cellular processes underlying addiction-related behaviours and propose novel hypotheses for the mechanisms by which orexin signalling may impart drug seeking. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Corey Baimel
- Department of Physiology and Pharmacology, The University of CalgaryCalgary, AB, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British ColumbiaVancouver, BC, Canada
| | - Selena E Bartlett
- Translational Research Institute, Institute for Health and Biomedical Sciences, Faculty of Health Queensland University of TechnologyBrisbane, QLD, Australia
| | - Lih-Chu Chiou
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan UniversityTaipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - John W Muschamp
- Center for Substance Abuse Research, Department of Pharmacology, School of Medicine, Temple UniversityPhiladelphia, PA, USA
| | - Omkar Patkar
- Translational Research Institute, Institute for Health and Biomedical Sciences, Faculty of Health Queensland University of TechnologyBrisbane, QLD, Australia
| | - Li-Wei Tung
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, The University of CalgaryCalgary, AB, Canada
| |
Collapse
|
50
|
López JM, Sanz-Morello B, González A. Organization of the orexin/hypocretin system in the brain of two basal actinopterygian fishes, the cladistians Polypterus senegalus and Erpetoichthys calabaricus. Peptides 2014; 61:23-37. [PMID: 25169954 DOI: 10.1016/j.peptides.2014.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/19/2014] [Accepted: 08/19/2014] [Indexed: 01/12/2023]
Abstract
Cladistians are primitive actinopterygian fishes mostly neglected in neuroanatomical studies. In the present study, the detailed neuroanatomical distribution of orexin (hypocretin)-like immunoreactive (OX-ir) cell bodies and fibers was analyzed in the brain of two species representative of the two extant genera of cladistians. Antibodies against mammalian orexin-A and orexin-B peptides were used. Simultaneous detection of orexins with neuropeptide Y (NPY), tyrosine hydroxylase (TH), and serotonin (5-HT) was used to establish accurately the topography of the orexin system and to evaluate the possible interactions with NPY and monoaminergic systems. A largely common pattern of OX-ir distribution in the two cladistian species was observed. Most OX-ir cells were located in the suprachiasmatic nucleus and tuberal hypothalamus, whereas scarce cells were observed in the posterior tubercle. In addition, a population of OX-ir cells was found in the preoptic area only in Polypterus and some cells also contained TH. The observed widespread distribution of OX-ir fibers was especially abundant in the retrobulbar area, subpallial areas, preoptic area, suprachiasmatic nucleus, tuberal hypothalamic area, prethalamus, thalamus, pretectum, optic tectum, and tegmentum. Low innervation was found in relation to monoaminergic cell groups, whereas a high NPY innervation was observed in all OX-ir cell groups. These relationships would represent the anatomical substrate for the functional interdependence between these systems. The organization of the orexin system in cladistians revealed a pattern largely consistent with those reported for all studied groups of vertebrates, suggesting that the primitive organization of this peptidergic system occurred in the common ancestor of gnathostome vertebrates.
Collapse
Affiliation(s)
- Jesús M López
- Department of Cell Biology, Faculty of Biology, University Complutense, 28040 Madrid, Spain
| | - Berta Sanz-Morello
- Department of Cell Biology, Faculty of Biology, University Complutense, 28040 Madrid, Spain
| | - Agustín González
- Department of Cell Biology, Faculty of Biology, University Complutense, 28040 Madrid, Spain.
| |
Collapse
|