1
|
Choi PP, Wang Q, Brenner LA, Li AJ, Ritter RC, Appleyard SM. Lesion of NPY Receptor-expressing Neurons in Perifornical Lateral Hypothalamus Attenuates Glucoprivic Feeding. Endocrinology 2024; 165:bqae021. [PMID: 38368624 PMCID: PMC11043786 DOI: 10.1210/endocr/bqae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/19/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Glucoprivic feeding is one of several counterregulatory responses (CRRs) that facilitates restoration of euglycemia following acute glucose deficit (glucoprivation). Our previous work established that glucoprivic feeding requires ventrolateral medullary (VLM) catecholamine (CA) neurons that coexpress neuropeptide Y (NPY). However, the connections by which VLM CA/NPY neurons trigger increased feeding are uncertain. We have previously shown that glucoprivation, induced by an anti-glycolygic agent 2-deoxy-D-glucose (2DG), activates perifornical lateral hypothalamus (PeFLH) neurons and that expression of NPY in the VLM CA/NPY neurons is required for glucoprivic feeding. We therefore hypothesized that glucoprivic feeding and possibly other CRRs require NPY-sensitive PeFLH neurons. To test this, we used the ribosomal toxin conjugate NPY-saporin (NPY-SAP) to selectively lesion NPY receptor-expressing neurons in the PeFLH of male rats. We found that NPY-SAP destroyed a significant number of PeFLH neurons, including those expressing orexin, but not those expressing melanin-concentrating hormone. The PeFLH NPY-SAP lesions attenuated 2DG-induced feeding but did not affect 2DG-induced increase in locomotor activity, sympathoadrenal hyperglycemia, or corticosterone release. The 2DG-induced feeding response was also significantly attenuated in NPY-SAP-treated female rats. Interestingly, PeFLH NPY-SAP lesioned male rats had reduced body weights and decreased dark cycle feeding, but this effect was not seen in female rats. We conclude that a NPY projection to the PeFLH is necessary for glucoprivic feeding, but not locomotor activity, hyperglycemia, or corticosterone release, in both male and female rats.
Collapse
Affiliation(s)
- Pique P Choi
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Qing Wang
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Lynne A Brenner
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Ai-Jun Li
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Robert C Ritter
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Suzanne M Appleyard
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
2
|
Elbaz BA, Fares I, Ahmed AM, Hegab IM. Different LED Light Colors Modify Behavior, Physiology, and Hypothalamic CRF and NPY mRNA Expression in Japanese Quail (Coturnix coturnix Japonica). Appl Anim Behav Sci 2023. [DOI: 10.1016/j.applanim.2023.105902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
3
|
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: 1.0] [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
|
4
|
Marcos P, Coveñas R. Involvement of the Orexinergic System in Feeding. APPLIED SCIENCES 2021; 12:86. [DOI: 10.3390/app12010086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
To know the processes involved in feeding, the dysregulation of hypothalamic neuropeptides promoting anorexigenic/orexigenic mechanisms must be investigated. Many neuropeptides are involved in this behavior and in overweight/obesity. Current pharmacological strategies for the treatment of obesity are unfortunately not very effective and, hence, new therapeutic strategies must be investigated and developed. Due to the crucial role played by orexins in feeding behavior, the aim of this review is to update the involvement of the orexinergic system in this behavior. The studies performed in experimental animal models and humans and the relationships between the orexinergic system and other substances are mentioned and discussed. Promising research lines on the orexinergic system are highlighted (signaling pathways, heterogeneity of the hypothalamic orexinergic neurons, receptor-receptor interaction, and sex differences). Each of the orexin 1 and 2 receptors plays a unique role in energy metabolism, exerting a differential function in obesity. Additional preclinical/clinical studies must be carried out to demonstrate the beneficial effects mediated by orexin receptor antagonists. Because therapies applied are in general ineffective when they are directed against a single target, the best option for successful anti-obesity treatments is the development of combination therapies as well as the development of new and more specific orexin receptor antagonists.
Collapse
Affiliation(s)
- Pilar Marcos
- CRIB (Regional Centre of Biomedical Research), Cellular Neuroanatomy and Molecular Chemistry of Central Nervous System, Faculty of Medicine, University of Castilla-La Mancha, Avenida de Almansa 14, 02006 Albacete, Spain
| | - Rafael Coveñas
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León (INCYL), University of Salamanca, c/Pintor Fernando Gallego 1, 37007 Salamanca, Spain
- Group GIR-BMD (Bases Moleculares del Desarrollo), University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
5
|
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: 4.0] [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
|
6
|
Yaeger JD, Krupp KT, Gale JJ, Summers CH. Counterbalanced microcircuits for Orx1 and Orx2 regulation of stress reactivity. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
7
|
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: 2.4] [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
|
8
|
Gumbs MCR, Eggels L, Kool T, Unmehopa UA, van den Heuvel JK, Lamuadni K, Mul JD, la Fleur SE. Neuropeptide Y Signaling in the Lateral Hypothalamus Modulates Diet Component Selection and is Dysregulated in a Model of Diet-Induced Obesity. Neuroscience 2019; 447:28-40. [PMID: 31887359 DOI: 10.1016/j.neuroscience.2019.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 01/16/2023]
Abstract
The preclinical multicomponent free-choice high-fat high-sucrose (fcHFHS) diet has strong validity to model diet-induced obesity (DIO) and associated maladaptive molecular changes in the central nervous system. fcHFHS-induced obese rats demonstrate increased sensitivity to intracerebroventricular infusion of the orexigenic Neuropeptide Y (NPY). The brain region-specific effects of NPY signaling on fcHFHS diet component selection are not completely understood. For example, fcHFHS-fed rats have increased intake of chow and fat following intracerebroventricular NPY infusion, whereas NPY administration in the nucleus accumbens, a key hub of the reward circuitry, specifically increases fat intake. Here, we investigated whether NPY infusion in the lateral hypothalamic area (LHA), which is crucially involved in the regulation of intake, regulates fcHFHS component selection, and if LHA NPY receptor subtypes 1 or 5 (NPYR1/5) are involved. Male Wistar rats were fed a chow or fcHFHS diet for at least seven days, and received intra-LHA vehicle or NPY infusions in a cross-over design. Diet component intake was measured two hours later. Separate experimental designs were used to test the efficacy of NPY1R- or NPY5R antagonism to prevent the orexigenic effects of intra-LHA NPY. Intra-LHA NPY increased caloric intake in chow- and fcHFHS-fed rats. This effect was mediated specifically by chow intake in fcHFHS-fed rats. The orexigenic effects of intra-LHA NPY were prevented by NPY1R and NPY5R antagonism in chow-fed rats, but only by NPY5R antagonism in fcHFHS-fed rats. Thus, NPY signaling has brain region-specific effects on fcHFHS component selection and LHA NPYR sensitivity is dysregulated during consumption of a fcHFHS diet.
Collapse
Affiliation(s)
- M C R Gumbs
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - L Eggels
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - T Kool
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - U A Unmehopa
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - J K van den Heuvel
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - K Lamuadni
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - J D Mul
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands
| | - S E la Fleur
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Intra-accumbal orexin-1 receptor inhibition prevents the anxiolytic-like effect of ethanol and leads to increases in orexin-A content and receptor expression. Pharmacol Biochem Behav 2019; 185:172761. [DOI: 10.1016/j.pbb.2019.172761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/23/2022]
|
10
|
Abstract
Despite decades of research, few medications have gained Food and Drug Administration (FDA) approval for the management of substance abuse disorder. The paucity of successful medications can be attributed, in part, to the lack of clearly identified neurobiological targets for addressing the core pathology of addictive behavior. Commonalities in the behavioral and brain processes involved in the rewarding effects of drugs and foods has prompted the evaluation of candidate medications that target neural pathways involved in both drug and eating disorders. Here, pharmacological strategies for the development of novel medications for drug addiction are presented in the context of potential overlapping neurobiological targets identified for eating disorders (e.g., obesity, overeating, binge-eating) and substance abuse. Mechanisms discussed in this chapter include modulators of the gut-brain axis (e.g., leptin, ghrelin, cholecystokinin, cocaine- and amphetamine-regulated transcript, and pancreatic peptides) and neurotransmitter systems (e.g., opioids, cannabinoids, dopamine, serotonin, and acetylcholine).
Collapse
|
11
|
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: 36] [Impact Index Per Article: 6.0] [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
|
12
|
Alvarez B, Barrientos T, Gac L, Teske J, Perez-Leighton C. Effects on Hedonic Feeding, Energy Expenditure and Balance of the Non-opioid Peptide DYN-A2-17. Neuroscience 2018; 371:337-345. [DOI: 10.1016/j.neuroscience.2017.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/11/2017] [Accepted: 11/27/2017] [Indexed: 11/28/2022]
|
13
|
Affiliation(s)
- I O Ebrahim
- Department of Psychiatry, Lane Fox Unit, St Thomas' Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | | | | | | | | |
Collapse
|
14
|
Sharko AC, Fadel JR, Kaigler KF, Wilson MA. Activation of orexin/hypocretin neurons is associated with individual differences in cued fear extinction. Physiol Behav 2017; 178:93-102. [PMID: 27746261 PMCID: PMC5391308 DOI: 10.1016/j.physbeh.2016.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/23/2016] [Accepted: 10/11/2016] [Indexed: 01/15/2023]
Abstract
Identifying the neurobiological mechanisms that underlie differential sensitivity to stress is critical for understanding the development and expression of stress-induced disorders, such as post-traumatic stress disorder (PTSD). Preclinical studies have suggested that rodents display different phenotypes associated with extinction of Pavlovian conditioned fear responses, with some rodent populations being resistant to extinction. An emerging literature also suggests a role for orexins in the consolidation processes associated with fear learning and extinction. To examine the possibility that the orexin system might be involved in individual differences in fear extinction, we used a Pavlovian conditioning paradigm in outbred Long-Evans rats. Rats showed significant variability in the extinction of cue-conditioned freezing and extinction recall, and animals were divided into groups based on their extinction profiles based on a median split of percent freezing behavior during repeated exposure to the conditioned cue. Animals resistant to extinction (high freezers) showed more freezing during repeated cue presentations during the within trial and between trial extinction sessions compared with the group showing significant extinction (low freezers), although there were no differences between these groups in freezing upon return to the conditioned context or during the conditioning session. Following the extinction recall session, activation of orexin neurons was determined using dual label immunohistochemistry for cFos in orexin positive neurons in the hypothalamus. Individual differences in the extinction of cue conditioned fear were associated with differential activation of hypothalamic orexin neurons. Animals showing poor extinction of cue-induced freezing (high freezers) had significantly greater percentage of orexin neurons with Fos in the medial hypothalamus than animals displaying significant extinction and good extinction recall (low freezers). Further, the freezing during extinction learning was positively correlated with the percentage of activated orexin neurons in both the lateral and medial hypothalamic regions. No differences in the overall density of orexin neurons or Fos activation were seen between extinction phenotypes. Although correlative, our results support other studies implicating a role of the orexinergic system in regulating extinction of conditioned responses to threat.
Collapse
Affiliation(s)
- Amanda C Sharko
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Jim R Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Kris F Kaigler
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Marlene A Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA.
| |
Collapse
|
15
|
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: 24] [Impact Index Per Article: 2.7] [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
|
16
|
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
|
17
|
Patel AX, Miller SR, Nathan PJ, Kanakaraj P, Napolitano A, Lawrence P, Koch A, Bullmore ET. Neuroendocrine and sympathetic responses to an orexin receptor antagonist, SB-649868, and alprazolam following insulin-induced hypoglycemia in humans. Psychopharmacology (Berl) 2014; 231:3817-28. [PMID: 24770625 PMCID: PMC4159598 DOI: 10.1007/s00213-014-3520-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/24/2014] [Indexed: 02/02/2023]
Abstract
RATIONALE The orexin-hypocretin system is important for translating peripheral metabolic signals and central neuronal inputs to a diverse range of behaviors, from feeding, motivation and arousal, to sleep and wakefulness. Orexin signaling is thus an exciting potential therapeutic target for disorders of sleep, feeding, addiction, and stress. OBJECTIVES/METHODS Here, we investigated the low dose pharmacology of orexin receptor antagonist, SB-649868, on neuroendocrine, sympathetic nervous system, and behavioral responses to insulin-induced hypoglycemic stress, in 24 healthy male subjects (aged 18-45 years; BMI 19.0-25.9 kg/m(2)), using a randomized, double-blind, placebo-controlled, within-subject crossover design. Alprazolam, a licensed benzodiazepine anxiolytic, was used as a positive comparator, as it has previously been validated using the insulin tolerance test (ITT) model in humans. RESULTS Of the primary endpoints, ITT induced defined increases in pulse rate, plasma cortisol, and adrenocorticotropic hormone in the placebo condition, but these responses were not significantly impacted by alprazolam or SB-649868 pre-treatment. Of the secondary endpoints, ITT induced a defined increase in plasma concentrations of adrenaline, noradrenaline, growth hormone (GH), and prolactin in the placebo condition. Alprazolam pre-treatment significantly reduced the GH response to ITT (p < 0.003), the peak electromyography (p < 0.0001) and galvanic skin response (GSR, p = 0.04) to acoustic startle, the resting GSR (p = 0.01), and increased appetite following ITT (p < 0.0005). SB-649868 pre-treatment produced no significant results. CONCLUSION We concluded that the ITT model may be informative for assessing the effects of drugs directly acting on the neuroendocrine or sympathetic nervous systems, but could not be validated for studying low dose orexin antagonist activity.
Collapse
Affiliation(s)
- Ameera X. Patel
- Brain Mapping Unit, Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK
| | - Sam R. Miller
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Pradeep J. Nathan
- Brain Mapping Unit, Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK ,Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK ,School of Psychology and Psychiatry, Monash University, Melbourne, Australia
| | - Ponmani Kanakaraj
- Quantitative Sciences India, GlaxoSmithKline Pharmaceuticals Ltd, Bangalore, India
| | - Antonella Napolitano
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Philip Lawrence
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Annelize Koch
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Edward T. Bullmore
- Brain Mapping Unit, Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK ,Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| |
Collapse
|
18
|
Wu WN, Wu PF, Zhou J, Guan XL, Zhang Z, Yang YJ, Long LH, Xie N, Chen JG, Wang F. Orexin-A activates hypothalamic AMP-activated protein kinase signaling through a Ca²⁺-dependent mechanism involving voltage-gated L-type calcium channel. Mol Pharmacol 2013; 84:876-87. [PMID: 24068427 DOI: 10.1124/mol.113.086744] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Hypothalamic AMP-activated protein kinase (AMPK) and orexins/hypocretins are both involved in the control of feeding behavior, but little is known about the interaction between these two signaling systems. Here, we demonstrated that orexin-A elicited significant activation of AMPK in the arcuate nucleus (ARC) of the hypothalamus by elevating cytosolic free Ca²⁺ involving extracellular calcium influx. Electrophysiological results revealed that orexin-A increased the L-type calcium current via the orexin receptor-phospholipase C-protein kinase C signaling pathway in ARC neurons that produce neuropeptide Y, an important downstream effector of orexin-A's orexigenic effect. Furthermore, the L-type calcium channel inhibitor nifedipine attenuated orexin-A-induced AMPK activation in vitro and in vivo. We found that inhibition of AMPK by either compound C (6-[4-[2-(1-piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)-pyrazolo[1,5-a]pyrimidine) or the ATP-mimetic 9-β-D-arabinofuranoside prevented the appetite-stimulating effect of orexin-A. This action can be mimicked by nifedipine, the blocker of the L-type calcium channel. Our results indicated that orexin-A activates hypothalamic AMPK signaling through a Ca²⁺-dependent mechanism involving the voltage-gated L-type calcium channel, which may serve as a potential target for regulating feeding behavior.
Collapse
Affiliation(s)
- Wen-Ning Wu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China (W.-N.W., P.-F.W., J.Z., X.-L.G., Z.Z., Y.-J.Y., L.-H.L., N.X., J.-G.C., F.W.); Key Laboratory of Neurologic Diseases (HUST), Ministry of Education of China, The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China (P.-F.W., L.-H.L., N.X., J.-G.C., F.W.); and Department of Pharmacology, Anhui Medical University, Hefei, China (W.-N.W.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Central administration of an orexin receptor 1 antagonist prevents the stimulatory effect of Olanzapine on endogenous glucose production. Brain Res 2013; 1527:238-45. [DOI: 10.1016/j.brainres.2013.06.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/18/2013] [Accepted: 06/25/2013] [Indexed: 12/31/2022]
|
20
|
Orexin-1 receptor antagonism fails to reduce anxiety-like behaviour in either plus-maze-naïve or plus-maze-experienced mice. Behav Brain Res 2013; 243:213-9. [PMID: 23333844 DOI: 10.1016/j.bbr.2012.12.064] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/13/2012] [Accepted: 12/17/2012] [Indexed: 11/20/2022]
Abstract
Although several lines of evidence have recently implicated orexins and their receptors in fear and anxiety, there is also a growing number of apparently inconsistent and/or negative findings. In the present study, we have used ethological methods to comprehensively profile the behavioural effects of the orexin-1 receptor antagonist SB-334867 (3-30 mg/kg) in mice exposed to the elevated plus-maze. Two experiments were performed, the first involving test-naïve animals and the second using prior undrugged experience of the maze to induce a qualitatively different emotional response to that seen on first exposure. In Experiment 1, a reference benzodiazepine (chlordiazepoxide, CDP, 15 mg/kg) produced a robust anxioselective profile comprising substantial increases in open arm exploration and reduced risk assessment without any signiifcant change in general activity levels. In contrast, SB-334867 failed to produce any behavioural effects over the dose range tested. In Experiment 2, 5 min undrugged experience of the maze 24h prior to testing increased open arm avoidance and abolished the anxiolytic efficacy of CDP. Despite this altered baseline, SB-334867 again failed to alter plus-maze behaviour. These findings agree with several recent reports that orexin receptor antagonists, such as SB-334867 and almorexant, do not alter basal anxiety levels in rats but markedly contrast with the anxiolytic-like effects of the same agents when anxiety levels have been exacerbated by fear conditioning, drug challenge or hypercapnia. This unique pattern of activity suggests that orexin receptor antagonists may have therapeutic value in those clinical anxiety disorders characterised by intense emotional arousal.
Collapse
|
21
|
Kermani M, Eliassi A. Gastric acid secretion induced by paraventricular nucleus microinjection of orexin A is mediated through activation of neuropeptide Yergic system. Neuroscience 2012; 226:81-8. [PMID: 22986171 DOI: 10.1016/j.neuroscience.2012.08.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/05/2012] [Accepted: 08/22/2012] [Indexed: 12/25/2022]
Abstract
UNLABELLED Very recently, we have reported that the modulatory effect of PVN on gastric acid secretion may be mediated through the orexin fibers and/or orexin-responsive neurons. In this study, we address the hypothesis which demonstrates the existence of a putative orexin A - neuropeptide Y Y1/Y5 receptors interaction to increase gastric acid secretion in pyloric-ligated conscious rats. Male Wistar rats were implanted with guide canula directed to the PVN and lateral ventricle. Intracerebroventricular (ICV) microinjections of GR-231118 (Y1 receptor antagonist) and CGP-71683 (Y5 receptor antagonist) on gastric acid secretion were considered. The effect of pretreatment with Y1 receptor antagonist, GR-231118, and Y5 receptor antagonist, CGP-71683, on PVN orexin A-induced acid secretion was assessed. Gastric acid secretion was measured using the pylorus-ligation method, and the amount of gastric acid was determined by titration with 0.01N NaOH to a pH of 7.0. KEY RESULTS ICV microinjections of GR-231118 and CGP-71683 decreased acid secretion by 25±0.05% and 67±0.02%, respectively. ICV microinjections of GR-231118 and CGP-71683 inhibited effects of PVN-injected orexin-A on acid secretion. We suggest that Y1 and Y5 receptors stimulate gastric acid secretion and the stimulatory effect of PVN orexin receptors on gastric acid secretion may be mediated via interactions, at least in part, through activation of Y1 and Y5 receptors. These neural pathways may play key roles in the orexinergic action of orexins in the cephalic phase of gastric acid secretion.
Collapse
Affiliation(s)
- M Kermani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
22
|
Nixon JP, Kotz CM, Novak CM, Billington CJ, Teske JA. Neuropeptides controlling energy balance: orexins and neuromedins. Handb Exp Pharmacol 2012:77-109. [PMID: 22249811 DOI: 10.1007/978-3-642-24716-3_4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this chapter, we review the feeding and energy expenditure effects of orexin (also known as hypocretin) and neuromedin. Orexins are multifunctional neuropeptides that affect energy balance by participating in regulation of appetite, arousal, and spontaneous physical activity. Central orexin signaling for all functions originates in the lateral hypothalamus-perifornical area and is likely functionally differentiated based on site of action and on interacting neural influences. The effect of orexin on feeding is likely related to arousal in some ways but is nonetheless a separate neural process that depends on interactions with other feeding-related neuropeptides. In a pattern distinct from other neuropeptides, orexin stimulates both feeding and energy expenditure. Orexin increases in energy expenditure are mainly by increasing spontaneous physical activity, and this energy expenditure effect is more potent than the effect on feeding. Global orexin manipulations, such as in transgenic models, produce energy balance changes consistent with a dominant energy expenditure effect of orexin. Neuromedins are gut-brain peptides that reduce appetite. There are gut sources of neuromedin, but likely the key appetite-related neuromedin-producing neurons are in the hypothalamus and parallel other key anorectic neuropeptide expression in the arcuate to paraventricular hypothalamic projection. As with other hypothalamic feeding-related peptides, hindbrain sites are likely also important sources and targets of neuromedin anorectic action. Neuromedin increases physical activity in addition to reducing appetite, thus producing a consistent negative energy balance effect. Together with the other various neuropeptides, neurotransmitters, neuromodulators, and neurohormones, neuromedin and orexin act in the appetite network to produce changes in food intake and energy expenditure, which ultimately influences the regulation of body weight.
Collapse
Affiliation(s)
- Joshua P Nixon
- Veterans Affairs Medical Center, Research Service (151), Minneapolis, MN, USA
| | | | | | | | | |
Collapse
|
23
|
López M, Tena-Sempere M, Diéguez C. Cross-talk between orexins (hypocretins) and the neuroendocrine axes (hypothalamic-pituitary axes). Front Neuroendocrinol 2010; 31:113-27. [PMID: 19654017 DOI: 10.1016/j.yfrne.2009.07.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 07/28/2009] [Accepted: 07/29/2009] [Indexed: 02/06/2023]
Abstract
Lesioning and electrical stimulation experiments carried out during the first half of the twentieth century showed that the lateral hypothalamic area (LHA) is involved in the neuroendocrine control of hormone secretion. However, the molecular basis of this phenomenon remained unclear until fifty years later when in 1998, two different laboratories discovered a new family of hypothalamic neuropeptides, the orexins or hypocretins (OX-A/Hcrt1 and OX-B/Hcrt2). Since then, remarkable evidence has revealed that orexins/hypocretins play a prominent role in regulating virtually all the neuroendocrine axes, acting as pivotal signals in the coordination of endocrine responses with regards to sleep, arousal and energy homeostasis. The clinical relevance of these actions is supported by human data showing impairment of virtually all the neuroendocrine axes in orexin/hypocretin-deficient narcoleptic patients. Here, we summarize more than ten years of knowledge about the orexins/hypocretins with particular focus on their role as neuroendocrine regulators. Understanding this aspect of orexin/hypocretin physiology could open new therapeutic possibilities in the treatment of sleep, energy homeostasis and endocrine pathologies.
Collapse
Affiliation(s)
- Miguel López
- Department of Physiology, School of Medicine, University of Santiago de Compostela - Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain.
| | | | | |
Collapse
|
24
|
López M, Nogueiras R, Tena-Sempere M, Diéguez C. Orexins (hypocretins) actions on the GHRH/somatostatin-GH axis. Acta Physiol (Oxf) 2010; 198:325-34. [PMID: 19769635 DOI: 10.1111/j.1748-1716.2009.02042.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The secretion of growth hormone (GH) is regulated through a complex neuroendocrine control system that includes two major hypothalamic regulators, namely GH-releasing hormone (GHRH) and somatostatin (SST) that stimulate and inhibit, respectively, GH release. Classical experiments involving damage and electrical stimulation suggested that the lateral hypothalamic area (LHA) modulated the somatotropic axis, but the responsible molecular mechanisms were unclear. Evidence obtained during the last decade has demonstrated that orexins/hypocretins, a family of peptides expressed in the LHA controlling feeding and sleep, play an important regulatory role on GH, by inhibiting its secretion modulating GHRH and SST neurones. Considering that GH release is closely linked to the sleep-wake cycle and feeding state, understanding orexin/hypocretin physiology could open new therapeutic possibilities in the treatment of sleep, energy homeostasis and GH-related pathologies, such as GH deficiency.
Collapse
Affiliation(s)
- M López
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.
| | | | | | | |
Collapse
|
25
|
Sharf R, Sarhan M, Dileone RJ. Role of orexin/hypocretin in dependence and addiction. Brain Res 2009; 1314:130-8. [PMID: 19699189 DOI: 10.1016/j.brainres.2009.08.028] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 07/30/2009] [Accepted: 08/01/2009] [Indexed: 01/08/2023]
Abstract
The orexins (or hypocretins) are hypothalamic neuropeptides that have been implicated in a variety of behaviors ranging from feeding to sleep and arousal. Evidence from animal models suggests a role for orexins in reward processing and drug addiction. In this review, we discuss orexin's interaction with the mesocorticolimbic reward pathway and the effects of drugs of abuse on the orexin system. We further review models of drug dependence and addiction and describe behavioral alterations that are seen when the orexin system is manipulated both pharmacologically and genetically. Based on the findings reported in the literature thus far, we posit that orexin functioning contributes to both drug reward and drug-related stress/aversive responsiveness; however, diverse anatomical substrates, and perhaps receptor specificity, contribute differentially to reward and stress components.
Collapse
Affiliation(s)
- Ruth Sharf
- Department of Psychiatry, Ribicoff Research Facilities, Yale University School of Medicine, New Haven, CT 06508, USA
| | | | | |
Collapse
|
26
|
Hirasawa M, Parsons MP, Alberto CO. Interaction between orexins and the mesolimbic system for overriding satiety. Rev Neurosci 2009; 18:383-93. [PMID: 19544624 DOI: 10.1515/revneuro.2007.18.5.383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In North American society, it is all too common for the intake of calories to outweigh an individual's energy demands. Such over-consumption where high-energy foods are readily available undoubtedly contributes to the growing problem of obesity. Palatable food stimulates brain circuits similar to those that mediate behavioral responses to drugs of abuse, which may underlie the continuation of food intake long after energy requirements are met. Among the brain areas implicated in reward and food intake, the lateral hypothalamus (LH) has long been recognized as a common region involved in both. It has been suggested that orexin neurons that are expressed exclusively within and adjacent to the LH comprise a major cellular substrate for the functioning of the LH. Here, we review the idea that the orexin neuropeptides play a key role in the rewarding aspects of food intake through interactions with both peripheral and central signals reflecting current energy stores as well as the classic reward pathway--the mesolimbic dopamine system. Furthermore, a possible heterogeneity of orexin neurons is discussed. Uncovering orexin's role in food reinforcement may provide insight into hyperphagia and obesity. In addition, the idea that food intake and substance abuse involve similar brain circuitry suggests potential for a single treatment aiding both obesity and addiction.
Collapse
Affiliation(s)
- Michiru Hirasawa
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada.
| | | | | |
Collapse
|
27
|
Solomon A, De Fanti BA, Martínez JA. Peripheral ghrelin interacts with orexin neurons in glucostatic signalling. ACTA ACUST UNITED AC 2007; 144:17-24. [PMID: 17619061 DOI: 10.1016/j.regpep.2007.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Ghrelin interactions with glycemia in appetite control as well as the potential mechanisms involving the orexin and melanin-concentrating hormone (MCH) neurons in the orexigenic ghrelin signals were investigated by using a specific anti-ghrelin antibody (AGA). Our results confirm that peripheral ghrelin is an important signal in meal initiation and appetite. Employing immunohistochemistry techniques, we found that c-fos positive neurons in the lateral hypothalamus (LH) and perifornical area (PFA) increased after insulin or 2-deoxyglucose administration. Moreover, we have also demonstrated that peripheral ghrelin blockade by the AGA, reduces the orexigenic signal induced by insulin and 2-DG administration probably partly producing a decrease of c-fos immunoreactivity in the LH and PFA as well as a lower activation of orexin neurons. In contrast, the c-fos positive MCH neurons were not apparently affected. In summary, our findings suggest that peripheral ghrelin plays an important role in regulatory "glucostatic" feeding mechanisms by means of its role as a "hunger" signal affecting the LH and PFA areas, which may contribute to energy homeostasis through orexin neurons.
Collapse
Affiliation(s)
- Andrew Solomon
- Department of Physiology and Nutrition, University of Navarra, 31008, Pamplona, Spain
| | | | | |
Collapse
|
28
|
Boutrel B, de Lecea L. Addiction and arousal: the hypocretin connection. Physiol Behav 2007; 93:947-51. [PMID: 18262574 DOI: 10.1016/j.physbeh.2007.11.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 11/02/2007] [Accepted: 11/02/2007] [Indexed: 11/26/2022]
Abstract
The hypocretins, also known as orexins, are two neuropeptides now commonly described as critical components to maintain and regulate the stability of arousal. Several lines of evidence have raised the hypothesis that hypocretin-producing neurons are part of the circuitries that mediate the hypothalamic response to acute stress. Intracerebral administration of hypocretin leads to a dose-related reinstatement of drug and food seeking behaviors. Furthermore, stress-induced reinstatement can be blocked with hypocretin receptor 1 antagonism. These results, together with recent data showing that hypocretin is critically involved in cocaine sensitization through the recruitment of NMDA receptors in the ventral tegmental area, strongly suggest that activation of hypocretin neurons play a critical role in the development of the addiction process. The activity of hypocretin neurons may affect addictive behavior by contributing to brain sensitization or by modulating the brain reward system. Hypocretinergic cells, in coordination with brain stress systems may lead to a vulnerable state that facilitates the resumption of drug seeking behavior. Hence, the hypocretinergic system is a new drug target that may be used to prevent relapse of drug seeking.
Collapse
Affiliation(s)
- Benjamin Boutrel
- Center for Psychiatric Neurosciences, Department of Psychiatry, University of Lausanne, Switzerland
| | | |
Collapse
|
29
|
Iwasa T, Matsuzaki T, Kiyokawa M, Shimizu F, Minakuchi M, Kuwahara A, Maegawa M, Yasui T, Irahara M. The type 2 corticotrophin-releasing hormone receptor mediates orexin A-induced luteinising hormone suppression in ovariectomised rats. J Neuroendocrinol 2007; 19:732-8. [PMID: 17680889 DOI: 10.1111/j.1365-2826.2007.01583.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Orexins are thought to be regulatory factors of the arousal and sleep patterns. They also affect immune, feeding, autonomic and neuroendocrine systems. We have previously shown that intracerebroventricular (i.c.v.) injection of orexin decreases pulsatile luteinising hormone (LH) secretion in ovariectomised (OVX) rats. However, the details of this mechanism have not been fully examined. Intracerebroventricular injection of orexin A also stimulates corticotrophin-releasing hormone (CRH) systems, which have been implicated in the stress-induced suppression of reproductive function. In the present study, we investigated the role of CRH systems in orexin-induced LH suppression. OVX rats were implanted with i.c.v. and intravenous (i.v.) cannulae. After i.c.v. injection of orexin and/or CRH receptor antagonists, blood samples were collected through the i.v. cannula at 6-min intervals for 120 min for LH measurement. Intracerebroventricular injection of orexin A or B (3 nmol/2.5 microl) suppressed pulsatile LH secretion. Coadministration of orexin A and alpha-helical corticotrophic-releasing factor (CRF), a nonselective CRH receptor antagonist (13 nmol/2.5 microl), or astressin(2)B, a selective type2 (CRH-R2) CRH receptor antagonist (28 nmol/2.5 microl), partly restored pulsatile LH secretion. Orexin B-induced LH suppression was not restored by alpha-helical CRF. In addition, i.c.v. injection of orexin A increased CRH and urocortin II (UcnII), but not Ucn mRNA levels, in the hypothalamus. These findings suggest that CRH-R2 mediates orexin A-induced LH suppression and it is possible that CRH and UcnII in the hypothalamus are involved in this pathway.
Collapse
Affiliation(s)
- T Iwasa
- Department of Obstetrics and Gynecology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
López M, Tovar S, Vázquez MJ, Williams LM, Diéguez C. Peripheral tissue-brain interactions in the regulation of food intake. Proc Nutr Soc 2007; 66:131-55. [PMID: 17343779 DOI: 10.1017/s0029665107005368] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
More than 70 years ago the glucostatic, lipostatic and aminostatic hypotheses proposed that the central nervous system sensed circulating levels of different metabolites, changing feeding behaviour in response to the levels of those molecules. In the last 20 years the rapid increase in obesity and associated pathologies in developed countries has involved a substantial increase in the knowledge of the physiological and molecular mechanism regulating body mass. This effort has resulted in the recent discovery of new peripheral signals, such as leptin and ghrelin, as well as new neuropeptides, such as orexins, involved in body-weight homeostasis. The present review summarises research into energy balance, starting from the original classical hypotheses proposing metabolite sensing, through peripheral tissue-brain interactions and coming full circle to the recently-discovered role of hypothalamic fatty acid synthase in feeding regulation. Understanding these molecular mechanisms will provide new pharmacological targets for the treatment of obesity and appetite disorders.
Collapse
Affiliation(s)
- Miguel López
- Department of Physiology, School of Medicine, University of Santiago de Compostela, C/San Francisco s/n 15782, Santiago de Compostela, A Coruña, Spain
| | | | | | | | | |
Collapse
|
31
|
Abstract
Spontaneous physical activity is activity that is non-volitional, or subconscious, such as fidgeting and shifting in one's seat, and time spent moving (standing and ambulating). Recent evidence indicates that spontaneous physical activity, and the resulting thermogenesis (non-exercise activity thermogenesis) may be regulated by brain systems. A large number of brain areas, with their associated neurotransmitter populations and connectivity, participate in the regulation of feeding behavior by acting as energy sensing and modulating centers. Although less well characterized, it is likely that a multitude of neurotransmitters and brain areas act to mediate spontaneous physical activity. These two behaviors, feeding and spontaneous physical activity, affect energy intake and expenditure and thus are important to body weight. Interestingly, often the two behaviors are affected simultaneously; when feeding is affected, so too is spontaneous physical activity, and both food intake and physical activity (whether spontaneous or volitional) influence activity of brain areas important to both. Several brain areas and neuropeptides are important to feeding and spontaneous physical activity. The lateral hypothalamus is one area that appears important to both behaviors, as stimulation or lesion of this region produces alterations in feeding behavior and spontaneous physical activity. Orexin neurons, with their central location in the lateral hypothalamus, widespread projections and connectivity to other brain areas important to energy homeostasis, are well situated to perform an integrative function. This review focuses on how hypothalamic orexins participate in both feeding and spontaneous physical activity, and provides potential models for the integration of signals important to both.
Collapse
Affiliation(s)
- Catherine M Kotz
- Veterans Affairs Medical Center, One Veterans Drive, GRECC (11G), Minneapolis, MN 55417, USA.
| |
Collapse
|
32
|
Winsky-Sommerer R, Boutrel B, de Lecea L. Stress and arousal: the corticotrophin-releasing factor/hypocretin circuitry. Mol Neurobiol 2006; 32:285-94. [PMID: 16385142 DOI: 10.1385/mn:32:3:285] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2005] [Accepted: 05/31/2005] [Indexed: 11/11/2022]
Abstract
The hypocretins (also know as orexins) are two neuropeptides now commonly described as critical components for maintaining and regulating the stability of arousal. Several lines of evidence have raised the hypothesis that hypocretin-producing neurons are part of the circuitries that mediate the hypothalamic response to acute stress. New data indicate that the corticotrophin-releasing factor (CRF) peptidergic system directly innervates hypocretin-expressing neurons. CRF depolarizes hypocretin neurons, and this effect is blocked by a CRF-R1 antagonist. Furthermore, activation of hypocretinergic neurons by stress is impaired in CRF-R1 knockout mice. These data suggest that CRF-R1 receptor mediates the stress-induced activation of the hypocretinergic system. A significant amount of evidence also indicates that hypocretin cells connect reciprocally to the CRF system. We propose that upon stressor stimuli, CRF activates the hypocretin system, which relays these signals to brain stem nuclei involved in the modulation of arousal as well as to the extended amygdala, a structure involved in the negative motivational state that drives addiction.
Collapse
|
33
|
Kokare DM, Patole AM, Carta A, Chopde CT, Subhedar NK. GABAA receptors mediate orexin-A induced stimulation of food intake. Neuropharmacology 2006; 50:16-24. [PMID: 16168444 DOI: 10.1016/j.neuropharm.2005.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2005] [Revised: 06/02/2005] [Accepted: 07/13/2005] [Indexed: 10/25/2022]
Abstract
Although the role of orexins in sleep/wake cycle and feeding behavior is well established, underlying mechanisms have not been fully understood. An attempt has been made to investigate the role of GABA(A) receptors and their benzodiazepine site on the orexin-A induced response to feeding. Different groups of rats were food deprived overnight and next day injected intracerebroventricularly (icv) with vehicle (artificial CSF; 5 microl/rat) or orexin-A (20-50 nM/rat) and the animals were given free access to food. Cumulative food intake was measured during light phase of light/dark cycle at 1-, 2-, 4- and 6-h post-injection time points. Orexin-A (30-50 nM/rat, icv) stimulated food intake at all the time points (P < 0.05). Prior administration of GABA(A) receptor agonists muscimol (25 ng/rat, icv) and diazepam (0.5 mg/kg, ip) at subeffective doses significantly potentiated the hyperphagic effect of orexin-A (30 nM/rat, icv). However, the effect was negated by the GABA(A) receptor antagonist bicuculline (1 mg/kg, ip). Interestingly, benzodiazepine receptor antagonist flumazenil (5 ng/rat, icv), augmented the orexin-A (30 nM/rat, icv) induced hyperphagia; the effect may be attributed to the intrinsic activity of the agent. The results suggest that the hyperphagic effect of orexin-A, at least in part, is mediated by enhanced GABA(A) receptor activity.
Collapse
|
34
|
Thorpe AJ, Cleary JP, Levine AS, Kotz CM. Centrally administered orexin A increases motivation for sweet pellets in rats. Psychopharmacology (Berl) 2005; 182:75-83. [PMID: 16075284 DOI: 10.1007/s00213-005-0040-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 04/24/2005] [Indexed: 11/24/2022]
Abstract
RATIONALE Centrally administered orexin A induces both feeding and locomotion in rats. Thus, the feeding response following orexin A administration may be secondary to general increases in activity rather than a specific motivation to eat. OBJECTIVE The aim of the study is to determine whether orexin A increases the motivation to eat. METHODS The effect of orexin A (0, 31.25, 62.5, 125, 250, and 500 pmol) on breakpoint was determined in male Sprague-Dawley rats with rostro-lateral hypothalamic cannulae under a progressive ratio of five schedule (PR5). The effect of orexin A (0, 31.25, 125, and 500 pmol) on pressing rate under a fixed ratio (20) schedule was obtained to analyze the time course of orexin-A-induced pressing. The effect of 24-h food deprivation on breakpoint under PR5 and the effect of orexin A (125 pmol) on free feeding (sweet pellets) and on open-field locomotor activity (0, 100, 500, and 1,000 pmol) were also tested. RESULTS Orexin A significantly augmented free feeding of sweet pellets, open-field locomotor activity, rate of pressing (FR20 schedule), and breakpoint (PR5 schedule), although compared to 24-h deprivation, the effect of orexin A on breakpoint was mild. However, there was a differential dose response relationship and time course of stimulation between orexin A's effects on locomotion and lever pressing. CONCLUSION These data indicate that infusion of orexin A enhances free feeding by enhancing and possibly prolonging motivation to eat.
Collapse
Affiliation(s)
- A J Thorpe
- Department of Neuroscience, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| | | | | | | |
Collapse
|
35
|
Ford GK, Al-Barazanji KA, Wilson S, Jones DNC, Harbuz MS, Jessop DS. Orexin expression and function: glucocorticoid manipulation, stress, and feeding studies. Endocrinology 2005; 146:3724-31. [PMID: 15961555 DOI: 10.1210/en.2005-0496] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the effects of glucocorticoid manipulation on orexin-A-induced feeding and prepro-orexin mRNA levels in the lateral hypothalamic area (LHA) of the rat brain. Adrenalectomy (ADX) reduced orexin-A-induced feeding over 4 h by about 60%, compared with shams, an effect that was reversed by corticosterone (CORT) replacement. ADX had no effect on prepro-orexin mRNA levels in the LHA in either the morning or the evening; however, message was up-regulated by CORT in the morning but not the evening. An increased number of emulsion grains per cell in the LHA suggests that this is a specific increase in prepro-orexin mRNA and is not due to an increased number of cells expressing message. Prepro-orexin mRNA levels in the LHA were elevated 4 h after injection of lipopolysaccharide, compared with saline-injected controls. Partial but not complete abolition of orexin-A-induced feeding by ADX suggests that orexin-A-induced feeding may be mediated through glucocorticoid-dependent and glucocorticoid-independent pathways. In the morning increased prepro-orexin mRNA after CORT replacement demonstrates that orexin expression is sensitive to increased concentrations of glucocorticoids. However, the lack of effect of ADX on prepro-orexin mRNA levels suggests that endogenous glucocorticoids are not involved in tonic regulation of basal prepro-orexin expression. Overall our data constitute a body of evidence for an integrated relationship between central orexin expression, stress, glucocorticoid manipulation, and feeding patterns in the rat.
Collapse
Affiliation(s)
- Gemma K Ford
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | | | | | | | | | | |
Collapse
|
36
|
Tebbe JJ, Mronga S, Tebbe CG, Ortmann E, Arnold R, Schäfer MKH. Ghrelin-induced stimulation of colonic propulsion is dependent on hypothalamic neuropeptide Y1- and corticotrophin-releasing factor 1 receptor activation. J Neuroendocrinol 2005; 17:570-6. [PMID: 16101895 DOI: 10.1111/j.1365-2826.2005.01340.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peptides participating in the hypothalamic control of feeding behaviour are also involved in the central autonomic control of gastrointestinal functions, such as secretion and motility. An anatomical interaction and functional relationship in the central nervous system between the feeding-related peptides neuropeptide Y and ghrelin is well documented. Furthermore, it has been shown that feeding-related peptides can influence digestive function via central corticotrophin-releasing factor (CRF) pathways. In the present study, we investigated the role of ghrelin in the central autonomic control of colonic motility. Furthermore, we addressed the hypothesis that ghrelin is involved in the hypothalamic control of colonic motor function, utilizing central neuropeptide Y receptors and hypothalamic CRF pathways. Ghrelin (0.03, 0.06 and 0.12 nmol) bilaterally microinjected into the paraventricular nucleus (PVN) induced a significant stimulation of colonic propulsion. In particular, the colonic transit time decreased from 312+/-7 min to 198+/-12 min. Microinjection of the neuropeptide Y1 receptor antagonist, BIBP-3226 (200 pmol), or the nonselective CRF receptor antagonist, astressin (30 pmol), into the PVN abolished the stimulatory effect of ghrelin injected into the PVN on colonic transit time, whereas pretreatment with the selective CRF2 receptor, antisauvagine-30 (28 pmol), failed to affect the effect of PVN-ghrelin injection on colonic propulsion. These results suggest that ghrelin can act as central modulator of gastrointestinal motor functions at the level of the PVN via neuropeptide Y1- and CRF1 receptor-dependent mechanisms.
Collapse
Affiliation(s)
- J J Tebbe
- Department of Internal Medicine, Division of Gastroenterology and Endocrinology, Philipps Universität Marburg, Marburg, Germany. ,de
| | | | | | | | | | | |
Collapse
|
37
|
Horowitz SS, Blanchard J, Morin LP. Medial vestibular connections with the hypocretin (orexin) system. J Comp Neurol 2005; 487:127-46. [PMID: 15880498 DOI: 10.1002/cne.20521] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The mammalian medial vestibular nucleus (MVe) receives input from all vestibular endorgans and provides extensive projections to the central nervous system. Recent studies have demonstrated projections from the MVe to the circadian rhythm system. In addition, there are known projections from the MVe to regions considered to be involved in sleep and arousal. In this study, afferent and efferent subcortical connectivity of the medial vestibular nucleus of the golden hamster (Mesocricetus auratus) was evaluated using cholera toxin subunit-B (retrograde), Phaseolus vulgaris leucoagglutinin (anterograde), and pseudorabies virus (transneuronal retrograde) tract-tracing techniques. The results demonstrate MVe connections with regions mediating visuomotor and postural control, as previously observed in other mammals. The data also identify extensive projections from the MVe to regions mediating arousal and sleep-related functions, most of which receive immunohistochemically identified projections from the lateral hypothalamic hypocretin (orexin) neurons. These include the locus coeruleus, dorsal and pedunculopontine tegmental nuclei, dorsal raphe, and lateral preoptic area. The MVe itself receives a projection from hypocretin cells. CTB tracing demonstrated reciprocal connections between the MVe and most brain areas receiving MVe efferents. Virus tracing confirmed and extended the MVe afferent connections identified with CTB and additionally demonstrated transneuronal connectivity with the suprachiasmatic nucleus and the medial habenular nucleus. These anatomical data indicate that the vestibular system has access to a broad array of neural functions not typically associated with visuomotor, balance, or equilibrium, and that the MVe is likely to receive information from many of the same regions to which it projects.
Collapse
Affiliation(s)
- Seth S Horowitz
- Department of Psychiatry, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | |
Collapse
|
38
|
Thorpe AJ, Teske JA, Kotz CM. Orexin A-induced feeding is augmented by caloric challenge. Am J Physiol Regul Integr Comp Physiol 2005; 289:R367-R372. [PMID: 15947069 DOI: 10.1152/ajpregu.00737.2004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Orexin neurons are stimulated by conditions that are glucoprivic, suggesting that orexin signaling may be increased during nutritional duress. We have previously shown that injection of orexin A (OxA) into the rostral lateral hypothalamic area (rLHa) robustly and dose-dependently increases feeding behavior. Thus we hypothesized that exogenous administration of orexin A would induce a greater feeding response after acute food deprivation or perceived caloric duress achieved through 2-deoxyglucose (2DG) administration. To test our hypothesis, male Sprague-Dawley rats implanted with internal guide cannulas directed to the rLHa were exposed to varying degrees of food deprivation (0, 3, 12, 24 h) and 2DG (200 mg/kg) before intra-rLHa OxA (500 pmol) infusion. We also performed a dose-response study using graded doses of OxA (0, 31.25, 125, and 500 pmol) in fed and 24-h fasted rats. OxA administration in conjunction with the highest level of prior food deprivation (24 h) resulted in the greatest feeding response (above baseline means; 0 h deprivation: 1.9 +/- 0.6; 24 h deprivation: 4.4 +/- 0.8; P = 0.0034) and showed a dose-dependent enhancement of feeding. Additionally, 2DG administration before OxA administration resulted in a significantly higher feeding response (above baseline means: 2DG = 1.8 +/- 0.5; OxA = 1.8 +/- 0.4; 2DG + OxA = 5.1 +/- 0.6; P < 0.0001). These data support the hypothesis that orexin signaling may be important in modulating the feeding network under times of nutritional duress.
Collapse
Affiliation(s)
- A J Thorpe
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | | | | |
Collapse
|
39
|
Fu LY, Acuna-Goycolea C, van den Pol AN. Neuropeptide Y inhibits hypocretin/orexin neurons by multiple presynaptic and postsynaptic mechanisms: tonic depression of the hypothalamic arousal system. J Neurosci 2005; 24:8741-51. [PMID: 15470140 PMCID: PMC6729969 DOI: 10.1523/jneurosci.2268-04.2004] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Neurons that release neuropeptide Y (NPY) have important effects on hypothalamic homeostatic regulation, including energy homeostasis, and innervate hypocretin neurons. Using whole-cell patch-clamp recording, we explored NPY actions on hypocretin cells identified by selective green fluorescent protein expression in mouse hypothalamic slices. NPY reduced spike frequency and hyperpolarized the membrane potential of hypocretin neurons. The NPY hyperpolarizing action persisted in tetrodotoxin (TTX), was mimicked by Y1 receptor-selective agonists [Pro34]-NPY and [D-Arg25]-NPY, and was abolished by the Y1-specific antagonist BIBP3226 [(R)-N2-(diphenylacetyl)-N-[(4-hydroxyphenyl)methyl]-D-arginine-amide], consistent with a direct activation of postsynaptic Y1 receptors. NPY induced a current that was dependent on extracellular potassium, reversed near the potassium equilibrium potential, showed inward rectification, was blocked by extracellular barium, and was abolished by GDP-betaS in the recording pipette, consistent with a G-protein-activated inwardly rectifying K+ (GIRK) current. [Pro34]-NPY evoked, and BIBP3226 blocked, the activation of the GIRK-type current, indicating mediation by a Y1 receptor. NPY attenuated voltage-dependent calcium currents mainly via a Y1 receptor subtype. BIBP3226 increased spontaneous spike frequency, suggesting an ongoing Y1 receptor-mediated NPY inhibition. In TTX, miniature EPSCs were reduced in frequency but not amplitude by NPY, NPY13-36, and [D-Trp32]-NPY, but not by [Pro34]-NPY, suggesting the presynaptic inhibition was mediated by a Y2/Y5 receptor. NPY had little effect on GABA-mediated miniature IPSCs but depressed spontaneous IPSCs. Together, these data support the view that NPY reduces the activity of hypocretin neurons by multiple presynaptic and postsynaptic mechanisms and suggest NPY axons innervating hypocretin neurons may tonically attenuate hypocretin-regulated arousal.
Collapse
Affiliation(s)
- Li-Ying Fu
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
40
|
Moreno G, Perelló M, Gaillard RC, Spinedi E. Orexin a stimulates hypothalamic-pituitary-adrenal (HPA) axis function, but not food intake, in the absence of full hypothalamic NPY-ergic activity. Endocrine 2005; 26:99-106. [PMID: 15888921 DOI: 10.1385/endo:26:2:099] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 02/11/2005] [Accepted: 02/22/2005] [Indexed: 11/11/2022]
Abstract
Neonatal monosodium L-glutamate (MSG) treatment destroys hypothalamic arcuate nucleus neuronal bodies, thus inducing several metabolic abnormalities. As a result, rats develop a phenotype characterized by hyperleptinemia and by impaired NPY but normal preproorexin hypothalamic mRNAs expression. Thus, our study was designed to explore whether hypothalamic effects of orexin A on food intake and glucocorticoid production develop in the absence of full hypothalamic NPY-ergic activity. For this purpose we evaluated, in control and MSG-treated rats, the consequences of intracerebroventricular (icv) orexin A administration on food intake and changes in circulating levels of ACTH and glucocorticoid. Our results indicate that orexin A icv treatment stimulated hypothalamic-pituitary-adrenal (HPA) axis activity in both MSG-damaged and normal animals, with this response even more pronounced in neurotoxin-damaged rats. Conversely, food intake was only enhanced by icv orexin A injection in normal rats. Our study further supports that acute hypothalamic effects of orexin A on food intake and glucocorticoid production are due to independent neuronal systems. While intact arcuate nucleus activity is needed for the orexinergic effect induced by icv orexin A administration, conversely, orexin A-stimulated HPA axis function takes place even in the absence of full NPY-ergic activity.
Collapse
Affiliation(s)
- Griselda Moreno
- Neuroendocrine Unit, Multidisciplinary Institute on Cell Biology (CONICET-CICPBA), 1900 La Plata, Argentina
| | | | | | | |
Collapse
|
41
|
Jørgensen EA, Knigge U, Watanabe T, Warberg J, Kjaer A. Histaminergic neurons are involved in the orexigenic effect of orexin-A. Neuroendocrinology 2005; 82:70-7. [PMID: 16415597 DOI: 10.1159/000090982] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Accepted: 11/04/2005] [Indexed: 11/19/2022]
Abstract
Orexin-A is an orexigenic peptide expressed mainly in the hypothalamus. Orexin-A increases and anti-orexin-A antibodies decrease food intake. However, the exact mechanism by which orexin-A exerts its orexigenic action is not fully elucidated. The histaminergic system is known to play a role in feeding behavior and we hypothesized that it could be involved in the orexigenic effect of orexin-A. To study this, we used histamine knockout animals and pharmacological blockade of the histaminergic system and studied the effect of orexin-A on feeding behavior and gene expression of neuropeptide Y (NPY). Orexin-A was administered intracerebroventricularly and food intake measured in wild-type, histamine H(1)-receptor knockout or histidine decarboxylase knockout mice. Additionally, we administered orexin-A to wild-type mice with pharmacologically blocked H(1)-receptors or pharmacologically stimulated autoinhibitory H(3)-receptors. By quantitative real-time PCR we measured the effect of orexin-A on NPY mRNA expression in wild-type and knockout mice. Orexin-A dose-dependently stimulated food intake when administered to wild-type mice in doses up to 0.03 microg. Orexin-A in a dose of 0.01 microg increased food intake 10-fold in wild-type mice, whereas no increase in food intake was seen in either knockout mice or pharmacologically manipulated mice. Orexin-A increased NPY mRNA 4-fold in wild-type mice, whereas no change was observed in knockout mice. We conclude that the orexigenic effect of orexin-A is dependent on an intact histaminergic neuronal system and seems to involve an H(1)-receptor mechanism.
Collapse
Affiliation(s)
- E A Jørgensen
- Department of Medical Physiology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | | | | | | | | |
Collapse
|
42
|
Winsky-Sommerer R, Yamanaka A, Diano S, Borok E, Roberts AJ, Sakurai T, Kilduff TS, Horvath TL, de Lecea L. Interaction between the corticotropin-releasing factor system and hypocretins (orexins): a novel circuit mediating stress response. J Neurosci 2004; 24:11439-48. [PMID: 15601950 PMCID: PMC6730356 DOI: 10.1523/jneurosci.3459-04.2004] [Citation(s) in RCA: 364] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 11/04/2004] [Accepted: 11/10/2004] [Indexed: 12/31/2022] Open
Abstract
The hypothalamic neuropeptides hypocretins (orexins) play a crucial role in the stability of arousal and alertness. We tested whether the hypocretinergic system is a critical component of the stress response activated by the corticotropin-releasing factor (CRF). Our results show that CRF-immunoreactive terminals make direct contact with hypocretin-expressing neurons in the lateral hypothalamus and that numerous hypocretinergic neurons express the CRF-R1/2 receptors. We also demonstrate that application of CRF to hypothalamic slices containing identified hypocretin neurons depolarizes membrane potential and increases firing rate in a subpopulation of hypocretinergic cells. CRF-induced depolarization was tetrodotoxin insensitive and was blocked by the peptidergic CRF-R1 antagonist astressin. Moreover, activation of hypocretinergic neurons in response to acute stress was severely impaired in CRF-R1 knock-out mice. Together, our data provide evidence of a direct neuroanatomical and physiological input from CRF peptidergic system onto hypocretin neurons. We propose that, after stressor stimuli, CRF stimulates the release of hypocretins and that this circuit contributes to activation and maintenance of arousal associated with the stress response.
Collapse
|
43
|
Sweet DC, Levine AS, Kotz CM. Functional opioid pathways are necessary for hypocretin-1 (orexin-A)-induced feeding. Peptides 2004; 25:307-14. [PMID: 15063013 DOI: 10.1016/j.peptides.2003.12.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2003] [Accepted: 12/22/2003] [Indexed: 11/18/2022]
Abstract
We investigated the interaction of the orexigenic neuropeptide, hypocretin-1 (Hcrt-1, also known as orexin-A), with endogenous opioids (also orexigenic neuropeptides). Rats were injected with naltrexone (NTX, nonspecific opioid antagonist) i.p., i.c.v., in the lateral hypothalamus (LH), and in the accumbens shell (AcbSh), and naloxone methiodide (nonspecific opioid antagonist unable to cross the blood brain barrier) was injected i.p. Rats were then injected with Hcrt-1 in the LH. Food intake was measured for up to 4h thereafter. Rats were also pretreated with NTX in the LH, with Hcrt-1 injected in the AcbSh. NTX suppressed Hcrt-1-induced feeding only when injected i.p., i.c.v., and in the AcbSh. These studies reveal the necessity for functional central opioidergic pathways involving the AcbSh, but not the LH in Hcrt-1-induced feeding.
Collapse
|
44
|
Abstract
Leptin, the long-sought satiety factor of adipocytes origin, has emerged as one of the major signals that relay the status of fat stores to the hypothalamus and plays a significant role in energy homeostasis. Understanding the mechanisms of leptin signaling in the hypothalamus during normal and pathological conditions, such as obesity, has been the subject of intensive research during the last decade. It is now established that leptin action in the hypothalamus in regulation of food intake and body weight is mediated by a neural circuitry comprising of orexigenic and anorectic signals, including NPY, MCH, galanin, orexin, GALP, alpha-MSH, NT, and CRH. In addition to the conventional JAK2-STAT3 pathway, it has become evident that PI3K-PDE3B-cAMP pathway plays a critical role in leptin signaling in the hypothalamus. It is now established that central leptin resistance contributes to the development of diet-induced obesity and ageing associated obesity. Central leptin resistance also occurs due to hyperleptinimia produced by exogenous leptin infusion. A defective nutritional regulation of leptin receptor gene expression and reduced STAT3 signaling may be involved in the development of leptin resistance in DIO. However, leptin resistance in the hypothalamic neurons may occur despite an intact JAK2-STAT3 pathway of leptin signaling. Thus, in addition to defective JAK2-STAT3 pathway, defects in other leptin signaling pathways may be involved in leptin resistance. We hypothesize that defective regulation of PI3K-PDE3B-cAMP pathway may be one of the mechanisms behind the development of central leptin resistance seen in obesity.
Collapse
Affiliation(s)
- Abhiram Sahu
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, S829 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
45
|
Chen C, Xu R. The in vitro regulation of growth hormone secretion by orexins. Endocrine 2003; 22:57-66. [PMID: 14610299 DOI: 10.1385/endo:22:1:57] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2003] [Accepted: 08/04/2003] [Indexed: 11/11/2022]
Abstract
Orexins, orexigenic neuropeptides, have recently been discovered in lateral hypothalamus and play an important role in the regulation of pituitary hormone secretion. Two subtypes of orexin receptors (orexin-1 and orexin-2) have been demonstrated in pituitaries. In this experiment, the effects of orexins on voltage-gated Ca2+ currents and the GH release in primary cultured ovine somatotropes were examined. Voltage-gated Ca2+ currents were isolated in ovine somatotropes as L, T, and N currents using whole-cell patch-clamp techniques and specific Ca2+ channel blocker and toxin. Application of orexin-A or orexin-B (100 nM) significantly, dose-dependently, and reversibly increased only nifedipine-sensitive L-type Ca2+ current. Inhibitors of PKC (calphostin C, PKC inhibitory peptide) but not inhibitors of PKA (H89, PKA inhibitory peptide) cancelled the increase in the L current by orexins. Co-administration of orexin-A and GHRH (10 nM) showed an additive effect on the L current. Specific intracellular Ca2+-store-depleting reagent, thapsigargin (1 microM), did not affect the orexin-induced increase in the L current. Orexin-B alone slightly increased GH release and co-administration of orexin-A and GHRH synergistically stimulated GH secretion in vitro. It is therefore suggested that orexins may play an important role in regulating GHRH-stimulated GH secretion through an increase in the L-type Ca2+ current and the PKC-mediated signaling pathways in ovine somatotropes.
Collapse
Affiliation(s)
- Chen Chen
- Prince Henry's Institute of Medical Research, and Department of Physiology, PO Box 5152, Monash University, Clayton, Victoria 3168, Australia.
| | | |
Collapse
|
46
|
Thorpe AJ, Mullett MA, Wang C, Kotz CM. Peptides that regulate food intake: regional, metabolic, and circadian specificity of lateral hypothalamic orexin A feeding stimulation. Am J Physiol Regul Integr Comp Physiol 2003; 284:R1409-17. [PMID: 12736178 DOI: 10.1152/ajpregu.00344.2002] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Orexin A (OX-A) administered in the lateral hypothalamus (LH) increases feeding in a dose-dependent manner. The LH is a relatively large neural structure with a heterogeneous profile of neural inputs, efferent projections, and orexin receptor distribution. We sought to determine the LH region most sensitive to the feeding stimulatory effect of OX-A injection. Fifty-six male Sprague-Dawley rats were fitted with cannulas 1 mm above four separate LH regions approximately 1 mm apart in the rostral-caudal direction. There were 14-16 animals/LH region. After recovery, animals received either artificial cerebrospinal fluid or OX-A (250, 500, or 1,000 pmol). To determine whether there is a circadian effect of LH OX-A on the feeding response, we performed injections at 0200, 0900, 1400, and 2100. Food intake was measured at 1, 2, and 4 h after injection. The most rostral extent of the LH was the only region in which injection of OX-A significantly stimulated feeding. Within this region, feeding was increased at all times of the day, although the most robust and only significant feeding response occurred after the afternoon injection (1400) of OX-A. To determine the extent to which the metabolic status of the rat contributed to the circadian specificity of orexin-induced feeding, animals were placed on a restricted diet and injected with OX-A in the most rostral region of the LH. Under these conditions, OX-A significantly increased feeding and more robustly when compared with animals on a nonrestricted diet. These data suggest that the rostral LH is the only region of the LH sensitive to the injection of OX-A, and the metabolic status of the animal at the time of injection may influence the feeding response to OX-A.
Collapse
Affiliation(s)
- Andrew J Thorpe
- Veterans Affairs Medical Center and Minnesota Obesity Center, Minneapolis 55417, USA
| | | | | | | |
Collapse
|
47
|
Xu R, Wang Q, Yan M, Hernandez M, Gong C, Boon WC, Murata Y, Ueta Y, Chen C. Orexin-A augments voltage-gated Ca2+ currents and synergistically increases growth hormone (GH) secretion with GH-releasing hormone in primary cultured ovine somatotropes. Endocrinology 2002; 143:4609-19. [PMID: 12446588 DOI: 10.1210/en.2002-220506] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Orexins are recently discovered neuropeptides that play an important role in the regulation of hormone secretion, and their receptors have been recently demonstrated in the pituitary. The effects of orexin-A on voltage-gated Ca2+ currents and GH release in primary cultured ovine somatotropes were examined. The expression of orexin-1 receptor was demonstrated by RT-PCR in ovine somatotropes, from which Ca2+ currents were also isolated as L, T, and N currents. Application of orexin-A (100 nM) significantly and reversibly increased only the L current, and coadministration of orexin-A and GHRH (10 nM) showed an additive effect on this current, but no effect of orexin-A was observed on either T or N current. Furthermore, the orexin-A-induced increase in the L current was completely abolished by the inhibition of protein kinase C (PKC) activity using calphostin C (100 nM), phorbal 12,13-dibutyrate pretreatment (0.5 micro M) for 16 h or specific PKC inhibitory peptide PKC(19-36) (1 mM). However, the increase in L current by orexin-A was sustained when cells were preincubated with a specific protein kinase A blocker H89 (1 micro M) or a specific intracellular Ca2+ store depleting reagent thapsigargin (1 micro M). Finally, orexin-A alone did not significantly increase GH release, but coadministration of orexin-A and GHRH showed a synergistic effect on GH secretion in vitro. Our results therefore suggest that orexin-A may play an important role in regulating GHRH-stimulated GH secretion through the enhancement of the L-type Ca2+ current and the PKC-mediated signaling pathway in ovine somatotropes.
Collapse
Affiliation(s)
- Ruwei Xu
- Prince Henry's Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Guan JL, Uehara K, Lu S, Wang QP, Funahashi H, Sakurai T, Yanagizawa M, Shioda S. Reciprocal synaptic relationships between orexin- and melanin-concentrating hormone-containing neurons in the rat lateral hypothalamus: a novel circuit implicated in feeding regulation. Int J Obes (Lond) 2002; 26:1523-32. [PMID: 12461668 DOI: 10.1038/sj.ijo.0802155] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2002] [Revised: 05/17/2002] [Accepted: 06/19/2002] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Both orexin (ORX)- and melanin-concentrating hormone (MCH) are expressed in different neurons in the lateral hypothalamic area (LH), and are considered to have common effects on stimulating food intake. There are no reports to demonstrate neural interactions between them at the ultrastructural level. We observed these neurons in the LH to evaluate the relationships between them. DESIGN We used two different types of double immunostaining to reveal the ultrastructure of both the ORX- and MCH-containing neurons. A preembedding double immunostaining technique was used to study the synaptic relationships between the two kinds of neuron. RESULTS The main new findings are as follows: 1) Both ORX- and MCH-containing neurons received other synaptic input and made synaptic input to other neurons; 2) Reciprocal synaptic relationships were observed between the ORX- and MCH-containing neurons. CONCLUSION The ORX- and MCH-containing neurons in the lateral hypothalamic area may influence food intake through synapse with each other.
Collapse
Affiliation(s)
- J-L Guan
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Initial research on the functional significance of two novel hypothalamic neuropeptides, orexin-A and orexin-B, suggested an important role in appetite regulation. Since then, however, these peptides have also been shown to influence a wide range of other physiological and behavioural processes. In this paper, we review the now quite extensive literature on orexins and appetite control, and consider their additional effects within this context. Although the evidence for orexin (particularly orexin-A and the orexin-1 receptor) involvement in many aspects of ingestive physiology and behaviour is incontrovertible, central administration of orexins is also associated with increased EEG arousal and wakefulness, locomotor activity and grooming, sympathetic and HPA activity, and pain thresholds. Since the orexin system is selectively activated by signals indicating severe nutritional depletion, it would be highly adaptive for a hungry animal not only to seek sustenance but also to remain fully alert to dangers in the environment. Crucial evidence indicates that orexin-A increases food intake by delaying the onset of a behaviourally normal satiety sequence. In contrast, a selective orexin-1 receptor antagonist (SB-334867) suppresses food intake and advances the onset of a normal satiety sequence. These data suggest that orexin-1 receptors mediate the episodic signalling of satiety and appear to bridge the transition from eating to resting in the rats' feeding-sleep cycle. The argument is developed that the diverse physiological and behavioural effects of orexins can best be understood in terms of an integrated set of reactions which function to rectify nutritional status without compromising personal survival. Indeed, many of the non-ingestive effects of orexin administration are identical to the cluster of active defences mediated via the lateral and dorsolateral columns of the midbrain periaqueductal gray matter, i.e., somatomotor activation, vigilance, tachycardia, hypertension and non-opioid analgesia. In our view, therefore, the LH orexin system is very well placed to orchestrate the diverse subsystems involved in foraging under potentially dangerous circumstances, i.e., finding and ingesting food without oneself becoming a meal for someone else.
Collapse
Affiliation(s)
- R J Rodgers
- School of Psychology, University of Leeds, Leeds, UK.
| | | | | | | |
Collapse
|
50
|
Stricker-Krongrad A, Beck B. Modulation of hypothalamic hypocretin/orexin mRNA expression by glucocorticoids. Biochem Biophys Res Commun 2002; 296:129-33. [PMID: 12147238 DOI: 10.1016/s0006-291x(02)00848-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The orexins are peptides which were recently isolated from the rat hypothlamus. They play a role in energy homeostasis and regulation of feeding as well as in other functions such as the sleep-wake cycle. The involvement of glucocorticoids in stress processes as well as in body weight regulation is well known. In the present paper, we investigated the role of glucocorticoids on hypocretin (Hcrt)/orexin (OX) pathway in Sprague-Dawley rats. We confirmed by in situ hybridization that prepro-Hcrt/OX mRNA expression is restricted to the lateral hypothalamus area with extension to the perifornical nucleus and the posterior hypothalamic area. Lateral hypothalamic prepro-Hcrt/OX mRNA expression was decreased by 50% after adrenalectomy (99.8+/-5.0 vs 49.2+/-4.4 nCi/g, p<0.01). Peripheral glucocorticoid treatment (dexamethasone) restored its expression to normal levels (105.4+/-6.1 nCi/g). The present data provide direct evidence that Hcrt/OX expression in the lateral hypothalamus is modulated by the glucocorticoids status. As the Hcrt/Ox system is closely interactive with the corticotropin-releasing hormone and neuropeptide Y systems, we propose that hypocretin/orexins peptides constitute a very sensitive key relay for mediating both stress and feeding behavior.
Collapse
Affiliation(s)
- Alain Stricker-Krongrad
- Metabolic Diseases Physiology and Pharmacology, Millennium Pharmaceuticals, 75 Sidney Street, MA 02139, Cambridge, USA
| | | |
Collapse
|