1
|
Zhuang C, Yan H, Lu J, Zhou Y, Liu Y, Shi G, Li Y. Compensatory enhancement of orexinergic system functionality induced by amyloid-β protein: a neuroprotective response in Alzheimer's disease. Front Physiol 2025; 16:1529981. [PMID: 40196718 PMCID: PMC11973307 DOI: 10.3389/fphys.2025.1529981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Background Amyloid-β protein (Aβ) accumulation is a defining characteristic of Alzheimer's disease (AD), resulting in neurodegeneration and a decline in cognitive function. Given orexin's well-documented role in enhancing memory and cognition, this study investigates its potential to regulate Aβ-induced neurotoxicity, offering new perspectives into AD management. Methods This paper simulated Aβ accumulation in the hippocampus of AD patients by administering Aβ1-42 oligomers into the bilateral hippocampal dentate gyrus of ICR mice. Inflammatory cytokines (IL-6, TNF-α) and orexin-A levels were measured by ELISA. Additionally, the excitability of orexinergic neurons was assessed by IHC targeting c-Fos expression. These methodologies evaluated the Aβ-induced neuroinflammation, orexinergic system functionality, and dexamethasone's (Dex) effects on these processes. Results Injection of Aβ1-42 oligomer resulted in elevated levels of IL-6, TNF-α, and orexin-A in the hippocampus, as well as increased excitability of orexinergic neurons in the lateral hypothalamus (LH). Dex treatment reduced neuroinflammation, causing a reduction in orexin-A levels and the excitability of orexinergic neurons. Conclusion Aβ-induced neuroinflammation is accompanied by enhanced levels of orexin-A and orexinergic neuron excitability. These findings suggest that the enhanced functionality of the orexinergic system may become a compensatory neuroprotective mechanism to counteract neuroinflammation and enhance cognitive function.
Collapse
Affiliation(s)
- Chenyu Zhuang
- Medical College, Yangzhou University, Yangzhou, China
| | - Hengyu Yan
- Medical College, Yangzhou University, Yangzhou, China
| | - Jiayu Lu
- Medical College, Yangzhou University, Yangzhou, China
| | - Yifan Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Yanqing Liu
- Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Guoshan Shi
- Department of Basic Medical Sciences, Guizhou University of Chinese Medicine, Guiyang, China
| | - Yan Li
- Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Department of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
2
|
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
|
3
|
Ameroso D, Rios M. Synaptic plasticity and the role of astrocytes in central metabolic circuits. WIREs Mech Dis 2024; 16:e1632. [PMID: 37833830 PMCID: PMC10842964 DOI: 10.1002/wsbm.1632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023]
Abstract
Neural circuits in the brain, primarily in the hypothalamus, are paramount to the homeostatic control of feeding and energy utilization. They integrate hunger, satiety, and body adiposity cues from the periphery and mediate the appropriate behavioral and physiological responses to satisfy the energy demands of the animal. Notably, perturbations in central homeostatic circuits have been linked to the etiology of excessive feeding and obesity. Considering the ever-changing energy requirements of the animal and required adaptations, it is not surprising that brain-feeding circuits remain plastic in adulthood and are subject to changes in synaptic strength as a consequence of nutritional status. Indeed, synapse density, probability of presynaptic transmitter release, and postsynaptic responses in hypothalamic energy balance centers are tailored to behavioral and physiological responses required to sustain survival. Mounting evidence supports key roles of astrocytes facilitating some of this plasticity. Here we discuss these synaptic plasticity mechanisms and the emerging roles of astrocytes influencing energy and glucose balance control in health and disease. This article is categorized under: Cancer > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
4
|
Braine A, Georges F. Emotion in action: When emotions meet motor circuits. Neurosci Biobehav Rev 2023; 155:105475. [PMID: 37996047 DOI: 10.1016/j.neubiorev.2023.105475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
The brain is a remarkably complex organ responsible for a wide range of functions, including the modulation of emotional states and movement. Neuronal circuits are believed to play a crucial role in integrating sensory, cognitive, and emotional information to ultimately guide motor behavior. Over the years, numerous studies employing diverse techniques such as electrophysiology, imaging, and optogenetics have revealed a complex network of neural circuits involved in the regulation of emotional or motor processes. Emotions can exert a substantial influence on motor performance, encompassing both everyday activities and pathological conditions. The aim of this review is to explore how emotional states can shape movements by connecting the neural circuits for emotional processing to motor neural circuits. We first provide a comprehensive overview of the impact of different emotional states on motor control in humans and rodents. In line with behavioral studies, we set out to identify emotion-related structures capable of modulating motor output, behaviorally and anatomically. Neuronal circuits involved in emotional processing are extensively connected to the motor system. These circuits can drive emotional behavior, essential for survival, but can also continuously shape ongoing movement. In summary, the investigation of the intricate relationship between emotion and movement offers valuable insights into human behavior, including opportunities to enhance performance, and holds promise for improving mental and physical health. This review integrates findings from multiple scientific approaches, including anatomical tracing, circuit-based dissection, and behavioral studies, conducted in both animal and human subjects. By incorporating these different methodologies, we aim to present a comprehensive overview of the current understanding of the emotional modulation of movement in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Anaelle Braine
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | | |
Collapse
|
5
|
Levine JA. The Fidget Factor and the obesity paradox. How small movements have big impact. Front Sports Act Living 2023; 5:1122938. [PMID: 37077429 PMCID: PMC10106700 DOI: 10.3389/fspor.2023.1122938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/24/2023] [Indexed: 04/05/2023] Open
Abstract
The hypothesis is that the Fidget Factor is the innate neurological pulse that propels humans and other species to move to support their health. Fidgets, previously thought to be spontaneous, are neurologically regulated and highly ordered (non-random). Modern societies being chair-based overwhelm Fidget Factor pulses and consequently inflict chair-based living for transportation, labor, and leisure. Despite impulses firing through the nervous system, people sit because environmental design overwhelms the biology. Urbanization and chair-based societies were designed after the industrial revolution to promote productivity; however, the consequence has been opposite. Crushing the natural urge to move—the Fidget Factor—is a public health calamity. Excess sitting is associated with a myriad of detrimental health consequences and impairs productivity. Fidgeting may reduce all-cause mortality associated with excessive sitting. The Fidget Factor offers hope; data demonstrate that workplaces and schools can be designed to promote activity and free people's Fidget Factors. Evidence shows that people are happier, healthier, wealthier, and more successful if their Fidget Factors are freed.
Collapse
|
6
|
Li Y, Guo Z, Cai C, Liu D, Kang Y, Liu P. The orexinergic system mediates the excitatory effects of caffeine on the arousal and sympathetic activity. Heliyon 2023; 9:e14170. [PMID: 36923861 PMCID: PMC10009538 DOI: 10.1016/j.heliyon.2023.e14170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Objective Caffeine is a non-selective adenosine receptor antagonist with pro-arousal and pro-sympathetic nervous system excitatory effects, and these pharmacological effects fit well with the physiological functions of orexin. The purpose of this study was to investigate the role of the orexinergic nervous system in the pharmacological effects of caffeine. Methods An animal model of sleepiness caused by adenosine accumulation was established by sleep deprivation, and caffeine's effects on the spontaneous activity and sympathetic nervous system of the model animals were evaluated by using the open-field experiment and gastrointestinal peristaltic observation, respectively, and the intervention of orexin receptor antagonists on the pharmacological effects of caffeine was also observed. Results Mice with 8 h of sleep deprivation showed a significant decrease in spontaneous activity and a significant increase in gastrointestinal push distance. After caffeine intervention, the spontaneous activities of sleep-deprived mice significantly increased and gastrointestinal peristalsis significantly decreased dose-dependent, while orexin receptors antagonist blocked the pro-arousal and inhibitory gastrointestinal peristalsis effects of caffeine on sleep-deprived mice. Conclusions Orexinergic nervous system mediated caffeine's excitatory effects on the pro-arousal and pro-sympathetic nervous systems. Orexin is likely to be an important performer in the pharmacological effects of caffeine.
Collapse
Affiliation(s)
- Yan Li
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, 225009, China.,Department of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, 225012, China
| | - Zhixuan Guo
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, 225009, China
| | - Chenxi Cai
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, 225009, China
| | - Danni Liu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, 225009, China
| | - Yin Kang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, 225009, China
| | - Pengfei Liu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, 225009, China
| |
Collapse
|
7
|
Gao XB, Horvath TL. From Molecule to Behavior: Hypocretin/orexin Revisited From a Sex-dependent Perspective. Endocr Rev 2022; 43:743-760. [PMID: 34792130 PMCID: PMC9277634 DOI: 10.1210/endrev/bnab042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 11/19/2022]
Abstract
The hypocretin/orexin (Hcrt/Orx) system in the perifornical lateral hypothalamus has been recognized as a critical node in a complex network of neuronal systems controlling both physiology and behavior in vertebrates. Our understanding of the Hcrt/Orx system and its array of functions and actions has grown exponentially in merely 2 decades. This review will examine the latest progress in discerning the roles played by the Hcrt/Orx system in regulating homeostatic functions and in executing instinctive and learned behaviors. Furthermore, the gaps that currently exist in our knowledge of sex-related differences in this field of study are discussed.
Collapse
Affiliation(s)
- Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
8
|
Villano I, La Marra M, Di Maio G, Monda V, Chieffi S, Guatteo E, Messina G, Moscatelli F, Monda M, Messina A. Physiological Role of Orexinergic System for Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8353. [PMID: 35886210 PMCID: PMC9323672 DOI: 10.3390/ijerph19148353] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023]
Abstract
Orexins, or hypocretins, are excitatory neuropeptides involved in the regulation of feeding behavior and the sleep and wakefulness states. Since their discovery, several lines of evidence have highlighted that orexin neurons regulate a great range of physiological functions, giving it the definition of a multitasking system. In the present review, we firstly describe the mechanisms underlining the orexin system and their interactions with the central nervous system (CNS). Then, the system's involvement in goal-directed behaviors, sleep/wakefulness state regulation, feeding behavior and energy homeostasis, reward system, and aging and neurodegenerative diseases are described. Advanced evidence suggests that the orexin system is crucial for regulating many physiological functions and could represent a promising target for therapeutical approaches to obesity, drug addiction, and emotional stress.
Collapse
Affiliation(s)
- Ines Villano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Marco La Marra
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Girolamo Di Maio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Vincenzo Monda
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80138 Naples, Italy; (V.M.); (E.G.)
| | - Sergio Chieffi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Ezia Guatteo
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80138 Naples, Italy; (V.M.); (E.G.)
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (G.M.); (F.M.)
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (G.M.); (F.M.)
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Antonietta Messina
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| |
Collapse
|
9
|
Mattar P, Uribe-Cerda S, Pezoa C, Guarnieri T, Kotz CM, Teske JA, Morselli E, Perez-Leighton C. Brain site-specific regulation of hedonic intake by orexin and DYN peptides: role of the PVN and obesity. Nutr Neurosci 2022; 25:1105-1114. [PMID: 33151127 DOI: 10.1080/1028415x.2020.1840049] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The orexin peptides promote hedonic intake and other reward behaviors through different brain sites. The opioid dynorphin peptides are co-released with orexin peptides but block their effects on reward in the ventral tegmental area (VTA). We previously showed that in the paraventricular hypothalamic nucleus (PVN), dynorphin and not orexin peptides enhance hedonic intake, suggesting they have brain-site-specific effects. Obesity alters the expression of orexin and dynorphin receptors, but whether their expression across different brain sites is important to hedonic intake is unclear. We hypothesized that hedonic intake is regulated by orexin and dynorphin peptides in PVN and that hedonic intake in obesity correlates with expression of their receptors. Here we show that in mice, injection of DYN-A1-13 (an opioid dynorphin peptide) in the PVN enhanced hedonic intake, whereas in the VTA, injection of OXA (orexin-A, an orexin peptide) enhanced hedonic intake. In PVN, OXA blunted the increase in hedonic intake caused by DYN-A1-13. In PVN, injection of norBNI (opioid receptor antagonist) reduced hedonic intake but a subsequent OXA injection failed to increase hedonic intake, suggesting that OXA activity in PVN is not influenced by endogenous opioid activity. In the PVN, DYN-A1-13 increased the intake of the less-preferred food in a two-food choice task. In obese mice fed a cafeteria diet, orexin 1 receptor mRNA across brain sites involved in hedonic intake correlated with fat preference but not caloric intake. Together, these data support that orexin and dynorphin peptides regulate hedonic intake in an opposing manner with brain-site-specific effects.
Collapse
Key Words
- CeA, central amygdala
- DH, dorsal hypothalamus
- DYN, dynorphin
- KOR, kappa opioid receptor
- LH, lateral hypothalamus
- NAc, nucleus accumbens
- OFC, orbitofrontal cortex
- OR, opioid receptor
- OX1R, orexin 1 receptor
- OX2R, orexin 2 receptor
- OXA, 1orexin-A
- Orexin
- PVN, paraventricular hypothalamic nucleus
- PVT, paraventricular thalamic nucleus
- VH, ventral hypothalamus
- VTA, ventral tegmental area
- cafeteria diet
- dynorphin
- fat
- feeding behavior
- food choice
- hedonic intake
- hypocretin
- hypothalamus
- norBNI, nor-binaltorphimine
- obesity
- opioid receptors
- orexin 1 receptor
Collapse
Affiliation(s)
- P Mattar
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Uribe-Cerda
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C Pezoa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - T Guarnieri
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C M Kotz
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - J A Teske
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - E Morselli
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C Perez-Leighton
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Mavanji V, Pomonis B, Kotz CM. Orexin, serotonin, and energy balance. WIREs Mech Dis 2022; 14:e1536. [PMID: 35023323 PMCID: PMC9286346 DOI: 10.1002/wsbm.1536] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022]
Abstract
The lateral hypothalamus is critical for the control of ingestive behavior and spontaneous physical activity (SPA), as lesion or stimulation of this region alters these behaviors. Evidence points to lateral hypothalamic orexin neurons as modulators of feeding and SPA. These neurons affect a broad range of systems, and project to multiple brain regions such as the dorsal raphe nucleus, which contains serotoninergic neurons (DRN) important to energy homeostasis. Physical activity is comprised of intentional exercise and SPA. These are opposite ends of a continuum of physical activity intensity and structure. Non‐goal‐oriented behaviors, such as fidgeting, standing, and ambulating, constitute SPA in humans, and reflect a propensity for activity separate from intentional activity, such as high‐intensity voluntary exercise. In animals, SPA is activity not influenced by rewards such as food or a running wheel. Spontaneous physical activity in humans and animals burns calories and could theoretically be manipulated pharmacologically to expend calories and protect against obesity. The DRN neurons receive orexin inputs, and project heavily onto cortical and subcortical areas involved in movement, feeding and energy expenditure (EE). This review discusses the function of hypothalamic orexin in energy‐homeostasis, the interaction with DRN serotonin neurons, and the role of this orexin‐serotonin axis in regulating food intake, SPA, and EE. In addition, we discuss possible brain areas involved in orexin–serotonin cross‐talk; the role of serotonin receptors, transporters and uptake‐inhibitors in the pathogenesis and treatment of obesity; animal models of obesity with impaired serotonin‐function; single‐nucleotide polymorphisms in the serotonin system and obesity; and future directions in the orexin–serotonin field. This article is categorized under:Metabolic Diseases > Molecular and Cellular Physiology
Collapse
Affiliation(s)
- Vijayakumar Mavanji
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Brianna Pomonis
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Catherine M Kotz
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA.,Geriatric Research Education and Clinical Center, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| |
Collapse
|
11
|
Aspesi D, Farinetti A, Marraudino M, Morgan GSK, Marzola E, Abbate-Daga G, Gotti S. Maternal separation alters the reward system of activity-based anorexia rats. Psychoneuroendocrinology 2021; 133:105393. [PMID: 34481327 DOI: 10.1016/j.psyneuen.2021.105393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
Maternal separation (MS) is a known chronic stressor in the postnatal period and when associated with another paradigm like the activity-based anorexia (ABA) rat model, causes different effects in the two sexes. In ABA females, the separation leads to increased hyperactivity and anxiety reduction, whereas, in males, the separation induces decreased locomotor activity without similar reduction of anxiety-like behaviors as observed in females. To understand the mechanisms altered by MS in synergy with the induction of the anorexic-like phenotype, we considered the reward system, which involves neurons synthesizing dopamine (DA) in the ventral tegmental area (VTA), substantia nigra pars compacta, and serotoninergic neurons in the dorsal raphe nucleus. Moreover, we analyzed the orexin circuit in the lateral hypothalamic area (LHA), which affects DA synthesis in the VTA and is also known to regulate food consumption and locomotor activity. Rats of both sexes were exposed to the two paradigms (MS and ABA), leading to four experimental groups for each sex: non-separated control (CON), non-separated ABA groups (ABA), MS control (MSCON), and MS plus ABA groups (MSABA). Immunohistochemistry analysis was performed to determine quantitative differences in the number of cells expressing DA, orexin, and serotonin (5-HT) among the experimental groups. The results showed that, in the DA system, the effect of MS was more evident in females than in males, with a substantial increase in DA cells in the VTA of MSABA. However, the analysis of the orexin system revealed a similar cellular increment in the LHA in the non-separated ABA groups of both sexes. Regarding 5-HT, there was an opposite effect in males and females of the MSABA groups, with only females showing a greater density of 5-HT cells. The changes in the reward system could partially explain the behavioral data: the hyperactivity, weight loss, and decreased anxiety levels of the MSABA females could be linked to an increase in DA and 5-HT cells, whereas in males, MS could mitigate the behavioral effects of the ABA protocol affecting the anxiety levels and locomotor activity through a lack of increased activation of the reward system.
Collapse
Affiliation(s)
- Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Ontario, N1G 2W1 Canada.
| | - Alice Farinetti
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy; NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, 10043 Turin, Italy.
| | - Marilena Marraudino
- NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, 10043 Turin, Italy.
| | - Godstime Stephen Kojo Morgan
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy; NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, 10043 Turin, Italy.
| | - Enrica Marzola
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy; Eating Disorders Center of the "Città della Salute e della Scienza" Hospital, University of Turin, Italy.
| | - Giovanni Abbate-Daga
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy; Eating Disorders Center of the "Città della Salute e della Scienza" Hospital, University of Turin, Italy.
| | - Stefano Gotti
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy; NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, 10043 Turin, Italy.
| |
Collapse
|
12
|
Smith LL. The Central Role of Hypothermia and Hyperactivity in Anorexia Nervosa: A Hypothesis. Front Behav Neurosci 2021; 15:700645. [PMID: 34421554 PMCID: PMC8377352 DOI: 10.3389/fnbeh.2021.700645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Typically, the development of anorexia nervosa (AN) is attributed to psycho-social causes. Several researchers have recently challenged this view and suggested that hypothermia and hyperactivity (HyAc) are central to AN. The following hypothesis will attempt to clarify their role in AN. Anorexia nervosa patients (ANs) have significantly lower core temperatures (Tcore) compared to healthy controls (HCs). This reduced temperature represents a reset Tcore that needs to be maintained. However, ANs cannot maintain this Tcore due primarily to a reduced basal metabolic rate (BMR); BMR usually supplies heat to sustain Tcore. Therefore, to generate the requisite heat, ANs revert to the behavioral-thermoregulatory strategy of HyAc. The majority of ANs (~89%) are reportedly HyAc. Surprisingly, engagement in HyAc is not motivated by a conscious awareness of low Tcore, but rather by the innocuous sensation of "cold- hands" frequently reported by ANs. That is, local hand-thermoreceptors signal the brain to initiate HyAc, which boosts perfusion of the hands and alters the sensation of "cold-discomfort" to one of "comfort." This "rewarding" consequence encourages repetition/habit formation. Simultaneously, hyperactivity increases the availability of heat to assist with the preservation of Tcore. Additionally, HyAc induces the synthesis of specific brain neuromodulators that suppress food intake and further promote HyAc; this outcome helps preserve low weight and perpetuates this vicious cycle. Based on this hypothesis and supported by rodent research, external heat availability should reduce the compulsion to be HyAc to thermoregulate. A reduction in HyAc should decrease the production of brain neuromodulators that suppress appetite. If verified, hopefully, this hypothesis will assist with the development of novel treatments to aid in the resolution of this intractable condition.
Collapse
Affiliation(s)
- Lucille Lakier Smith
- Human Performance Laboratory, Department of Kinesiology, School of Health Sciences, East Carolina University, Greenville, NC, United States
| |
Collapse
|
13
|
Olsen N, Furlong TM, Carrive P. Behavioural and cardiovascular effects of orexin-A infused into the central amygdala under basal and fear conditions in rats. Behav Brain Res 2021; 415:113515. [PMID: 34371088 DOI: 10.1016/j.bbr.2021.113515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 07/16/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
The neuropeptide orexin-A (OX-A) has diverse functions, including maintaining arousal, autonomic control, motor activity and stress responses. These functions are regulated at different terminal regions where OX-A is released. The current study examined the physiological and behavioural effects of OX-A microinjections into the central amygdala (CeA) under basal and stressed conditions in rats. When OX-A was microinjected into the CeA and the animals returned to the home-cage, heart rate and mean arterial pressure were increased compared to vehicle-injected controls. General activity of the animal was also increased, indicating that OX-A activity in CeA contributes to increased arousal. This outcome is similar to the effects of central intracerebroventricular infusions of OX-A, as well as the cardiovascular effects previously demonstrated at many of OX's efferent hypothalamic and brainstem structures. In a second study, animals were fear-conditioned to a context by delivery of electric footshocks and then animals were re-exposed to the conditioned context at test. When OX-A was microinjected at test, freezing behaviour was reduced and there was a corresponding increase in the animal's activity but no impact on the pressor and cardiac responses (i.e, blood pressure and heart rate were unchanged). This reduction in freezing suggests that OX-A activates amygdala neurons that inhibit freezing, which is similar to the actions of other neuropeptides in the CeA that modulate the appropriate defence response to fearful stimuli. Overall, these data indicate that the CeA is an important site of OX-A modulation of cardiovascular and motor activity, as well as conditioned freezing responses.
Collapse
Affiliation(s)
- Nick Olsen
- School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Teri M Furlong
- School of Medical Sciences, The University of New South Wales, Sydney, Australia; Neuroscience Research Australia, Randwick, Australia.
| | - Pascal Carrive
- School of Medical Sciences, The University of New South Wales, Sydney, Australia
| |
Collapse
|
14
|
Abstract
Twenty-two years after their discovery, the hypocretins (Hcrts), also known as orexins, are two of the most studied peptidergic systems, involved in myriad physiological systems that range from sleep, arousal, motivation, homeostatic regulation, fear, anxiety and learning. A causal relationship between activity of Hcrt and arousal stability was established shortly after their discovery and have led to the development of a new class of drugs to treat insomnia. In this review we discuss the many faces of the Hcrt system and examine recent findings that implicate decreased Hcrt function in the pathogenesis of a number of neuropsychiatric conditions. We also discuss future therapeutic strategies to replace or enhance Hcrt function as a treatment option for these neuropsychiatric conditions.
Collapse
Affiliation(s)
- Erica Seigneur
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
15
|
Orexins role in neurodegenerative diseases: From pathogenesis to treatment. Pharmacol Biochem Behav 2020; 194:172929. [PMID: 32315694 DOI: 10.1016/j.pbb.2020.172929] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Orexin is a neurotransmitter that mainly regulates sleep/wake cycle. In addition to its sleep cycle regulatory role, it is involved in regulation of attention, energy homeostasis, neurogenesis and cognition. Several evidences has shown the involvement of orexin in narcolepsy, but there are also growing evidences that shows the disturbance in orexin system in neurodegenerative diseases including Alzheimer's, Parkinson's, Epilepsy, Huntington's diseases and Amyotrophic lateral sclerosis. Pathogenesis and clinical symptoms of these disorders can be partly attributed from orexin system imbalance. However, there are controversial reports on the exact relationship between orexin and these neurodegenerative diseases. Therefore, the aim of this review is to summarize the current evidences regarding the role of orexin in these neurodegenerative diseases.
Collapse
|
16
|
Li SB, de Lecea L. The hypocretin (orexin) system: from a neural circuitry perspective. Neuropharmacology 2020; 167:107993. [PMID: 32135427 DOI: 10.1016/j.neuropharm.2020.107993] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| |
Collapse
|
17
|
Effects of DCM Leaf Extract of Gnidia glauca (Fresen) on Locomotor Activity, Anxiety, and Exploration-Like Behaviors in High-Fat Diet-Induced Obese Rats. Behav Neurol 2019; 2019:7359235. [PMID: 31933694 PMCID: PMC6942765 DOI: 10.1155/2019/7359235] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/03/2019] [Accepted: 12/07/2019] [Indexed: 11/30/2022] Open
Abstract
Obesity is the main component of metabolic syndromes involving distinct etiologies that target different underlying behavioral and physiological functions within the brain structures and neuronal circuits. An alteration in the neuronal circuitry stemming from abdominal or central obesity stimulates a cascade of changes in neurochemical signaling that directly or indirectly mediate spontaneously emitted behaviors such as locomotor activity patterns, anxiety, and exploration. Pharmacological agents available for the treatment of neurologic disorders have been associated with limited potency and intolerable adverse effects. These have necessitated the upsurge in the utilization of herbal prescriptions due to their affordability and easy accessibility and are firmly embedded within wider belief systems of many people. Gnidia glauca has been used in the management of many ailments including obesity and associated symptomatic complications. However, its upsurge in use has not been accompanied by empirical determination of these folkloric claims. The present study, therefore, is aimed at determining the modulatory effects of dichloromethane leaf extract of Gnidia glauca on locomotor activity, exploration, and anxiety-like behaviors in high-fat diet-induced obese rats in an open-field arena. Obesity was experimentally induced by feeding the rats with prepared high-fat diet and water ad libitum for 6 weeks. The in vivo antiobesity effects were determined by oral administration of G. glauca at dosage levels of 200, 250, and 300 mg/kg body weight in high-fat diet-induced obese rats from the 6th to 12th week. Phytochemical analysis was done using gas chromatography linked to mass spectroscopy. Results indicated that Gnidia glauca showed anxiolytic effects and significantly increased spontaneous locomotor activity and exploration-like behaviors in HFD-induced obese rats. The plant extract also contained phytocompounds that have been associated with amelioration of the main neurodegenerative mediators, viz., inflammation and oxidative stress. These findings provide “qualified leads” for the synthesis of new alternative therapeutic agents for the management of neurologic disorders. However, there is a need to conduct toxicity studies of Gnidia glauca to establish its safety profiles.
Collapse
|
18
|
Distinct effects of orexin receptor antagonist and GABA A agonist on sleep and physical/cognitive functions after forced awakening. Proc Natl Acad Sci U S A 2019; 116:24353-24358. [PMID: 31712421 DOI: 10.1073/pnas.1907354116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The majority of patients with insomnia are treated with hypnotic agents. In the present study, we evaluated the side-effect profile of an orexin receptor antagonist and γ-aminobutyric acid A (GABAA) receptor agonist on physical/cognitive functions upon forced awakening. This double-blind, randomized, placebo-controlled, cross-over study was conducted on 30 healthy male subjects. Fifteen minutes before bedtime, the subjects took a pill of suvorexant (20 mg), brotizolam (0.25 mg), or placebo and were forced awake 90 min thereafter. Physical- and cognitive-function tests were performed before taking the pill, after forced awakening, and the next morning. Polysomnographic recordings revealed that the efficacies of the hypnotic agents in prolonging total sleep time (∼30 min) and increasing sleep efficiency (∼6%) were comparable. When the subjects were allowed to go back to sleep after the forced awakening, the sleep latency was shorter under the influence of hypnotic agents (∼2 min) compared to the placebo trial (24 min), and the rapid eye movement latency was significantly shorter under suvorexant (98.8, 81.7, and 48.8 min for placebo, brotizolam, and suvorexant, respectively). Although brotizolam significantly impaired the overall physical/cognitive performance (sum of z score) compared with placebo upon forced awakening, there was no significant difference in the total z score of performance between suvorexant and placebo. Notably, the score for static balance with the eyes open was higher under suvorexant compared to brotizolam administration. The energy expenditure was lower under suvorexant and brotizolam compared with the placebo. The effect size of brotizolam (d = 0.24) to reduce the energy expenditure was larger than that of suvorexant (d < 0.01).
Collapse
|
19
|
Wang Y, Chen AQ, Xue Y, Liu MF, Liu C, Liu YH, Pan YP, Diao HL, Chen L. Orexins alleviate motor deficits via increasing firing activity of pallidal neurons in a mouse model of Parkinson's disease. Am J Physiol Cell Physiol 2019; 317:C800-C812. [PMID: 31365289 DOI: 10.1152/ajpcell.00125.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Orexin is a peptide neurotransmitter released in the globus pallidus. Morphological evidence reveals that both orexin 1 receptor (OX1R) and orexin 2 receptor (OX2R) exist in the globus pallidus. Here we showed that bilateral microinjection of both orexin-A and orexin-B into the globus pallidus alleviated motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian mice. Further in vivo extracellular single-unit recording revealed that the basal spontaneous firing rate of the globus pallidus neurons in MPTP parkinsonian mice was slower than that of normal mice. Application of orexin-A or orexin-B significantly increased the spontaneous firing rate of pallidal neurons. The influx of Ca2+ through the L-type Ca2+ channel is the major mechanism involved in orexin-induced excitation in the globus pallidus. Orexin-A-induced increase in firing rate of pallidal neurons in MPTP parkinsonian mice was stronger than that of normal mice. Orexin-A exerted both electrophysiological and behavioral effects mainly via OX1R, and orexin-B exerted the effects via OX2R. Endogenous orexins modulated the excitability of globus pallidus neurons mainly through OX1R. The present behavioral and electrophysiological results suggest that orexins ameliorate parkinsonian motor deficits through increasing the spontaneous firing of globus pallidus neurons.
Collapse
Affiliation(s)
- Ying Wang
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - An-Qi Chen
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Xue
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Mei-Fang Liu
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Cui Liu
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yun-Hai Liu
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yi-Peng Pan
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hui-Ling Diao
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Noise-induced sleep disruption increases weight gain and decreases energy metabolism in female rats. Int J Obes (Lond) 2018; 43:1759-1768. [PMID: 30568267 PMCID: PMC6584067 DOI: 10.1038/s41366-018-0293-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 01/28/2023]
Abstract
Background/objectives: Inadequate sleep increases obesity and environmental noise contributes to poor sleep. However, women may be more vulnerable to noise and hence more susceptible to sleep disruption-induced weight gain than men. In male rats, exposure to environmental (i.e. ambient) noise disrupts sleep and increases feeding and weight gain. However, the effects of environmental noise on sleep and weight gain in female rats are unknown. Thus, this study was designed to determine whether noise exposure would disturb sleep, increase feeding and weight gain and alter the length of the estrous cycle in female rats. Subjects/methods: Female rats (12-weeks old) were exposed to noise for 17d (8h/d during the light period) to determine the effects of noise on weight gain and food intake. In a separate set of females, estrous cycle phase and length, EEG, EMG, spontaneous physical activity and energy expenditure were recorded continuously for 27d during baseline (control, 9d), noise exposure (8h/d, 9d) and recovery (9d) from sleep disruption. Results: Noise exposure significantly increased weight gain and food intake compared to females that slept undisturbed. Noise also significantly increased wakefulness, reduced sleep and resulted in rebound sleep during the recovery period. Total energy expenditure was significantly lower during both noise exposure and recovery due to lower energy expenditure during spontaneous physical activity and sleep. Notably, noise did not alter the estrous cycle length. Conclusions: As previously observed in male rats, noise exposure disrupted sleep and increased weight gain in females but did not alter the length of the estrous cycle. This is the first demonstration of weight gain in female rats during sleep disruption. We conclude that the sleep disruption caused by exposure to environmental noise is a significant tool for determining how sleep loss contributes to obesity in females.
Collapse
|
21
|
Liu MF, Xue Y, Liu C, Liu YH, Diao HL, Wang Y, Pan YP, Chen L. Orexin-A Exerts Neuroprotective Effects via OX1R in Parkinson's Disease. Front Neurosci 2018. [PMID: 30524223 DOI: 10.3389/fnins.2018.00835.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by progressive and selective death of dopaminergic neurons. Orexin-A is involved in many biological effects of the body. It has been reported that orexin-A has protective effects in cellular models of PD. However, little is known about the protective effects of orexin-A in animal parkinsonian models and the cellular mechanism has not yet been fully clarified. The aim of this study was to evaluate the effects of orexin-A in MPTP mice model of PD as well as the possible neuroprotective mechanisms of orexin-A on dopaminergic neurons. The results from animal experiments demonstrated that orexin-A attenuated the loss of dopaminergic neurons and the decrease of tyrosine hydroxylase (TH) expression in the substantia nigra, normalized the striatal dopaminergic fibers, and prevented the depletion of dopamine and its metabolites in the striatum. MPTP-treated mice showed cognitive impairments accompanied with significant motor deficiency. Orexin-A improved MPTP-induced impairments in both motor activity and spatial memory. Importantly, orexin-A increased the protein level of brain-derived neurotrophic factor (BDNF) in dopaminergic neurons of the substantia nigra. Furthermore, the protective effects of orexin-A on MPTP parkinsonian mice could be blocked by orexinergic receptor 1 (OX1R) antagonist, SB334867. In another set of experiments with SH-SY5Y dopaminergic cells, orexin-A significantly induced the expression of BDNF in a dose and time-dependent manner. The upregulation of BDNF is mainly concerned with PI3K and PKC signaling pathways via OX1R. The present study demonstrated that orexin-A exerted neuroprotective effects on MPTP parkinsonian mice, which may imply orexin-A as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Mei-Fang Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China.,College of Pharmacy, Jining Medical University, Rizhao, China
| | - Yan Xue
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Cui Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Yun-Hai Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Hui-Ling Diao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Ying Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Yi-Peng Pan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Liu MF, Xue Y, Liu C, Liu YH, Diao HL, Wang Y, Pan YP, Chen L. Orexin-A Exerts Neuroprotective Effects via OX1R in Parkinson's Disease. Front Neurosci 2018; 12:835. [PMID: 30524223 PMCID: PMC6262320 DOI: 10.3389/fnins.2018.00835] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disorder characterized by progressive and selective death of dopaminergic neurons. Orexin-A is involved in many biological effects of the body. It has been reported that orexin-A has protective effects in cellular models of PD. However, little is known about the protective effects of orexin-A in animal parkinsonian models and the cellular mechanism has not yet been fully clarified. The aim of this study was to evaluate the effects of orexin-A in MPTP mice model of PD as well as the possible neuroprotective mechanisms of orexin-A on dopaminergic neurons. The results from animal experiments demonstrated that orexin-A attenuated the loss of dopaminergic neurons and the decrease of tyrosine hydroxylase (TH) expression in the substantia nigra, normalized the striatal dopaminergic fibers, and prevented the depletion of dopamine and its metabolites in the striatum. MPTP-treated mice showed cognitive impairments accompanied with significant motor deficiency. Orexin-A improved MPTP-induced impairments in both motor activity and spatial memory. Importantly, orexin-A increased the protein level of brain-derived neurotrophic factor (BDNF) in dopaminergic neurons of the substantia nigra. Furthermore, the protective effects of orexin-A on MPTP parkinsonian mice could be blocked by orexinergic receptor 1 (OX1R) antagonist, SB334867. In another set of experiments with SH-SY5Y dopaminergic cells, orexin-A significantly induced the expression of BDNF in a dose and time-dependent manner. The upregulation of BDNF is mainly concerned with PI3K and PKC signaling pathways via OX1R. The present study demonstrated that orexin-A exerted neuroprotective effects on MPTP parkinsonian mice, which may imply orexin-A as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Mei-Fang Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China.,College of Pharmacy, Jining Medical University, Rizhao, China
| | - Yan Xue
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Cui Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Yun-Hai Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Hui-Ling Diao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Ying Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Yi-Peng Pan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Liu C, Xue Y, Liu MF, Wang Y, Liu ZR, Diao HL, Chen L. Orexins increase the firing activity of nigral dopaminergic neurons and participate in motor control in rats. J Neurochem 2018; 147:380-394. [DOI: 10.1111/jnc.14568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Cui Liu
- Department of Physiology; Qingdao University; Qingdao China
| | - Yan Xue
- Department of Physiology; Qingdao University; Qingdao China
| | - Mei-Fang Liu
- Department of Physiology; Qingdao University; Qingdao China
| | - Ying Wang
- Department of Physiology; Qingdao University; Qingdao China
| | - Zi-Ran Liu
- Department of Physiology; Qingdao University; Qingdao China
| | - Hui-Ling Diao
- Department of Physiology; Qingdao University; Qingdao China
| | - Lei Chen
- Department of Physiology; Qingdao University; Qingdao China
| |
Collapse
|
24
|
|
25
|
Barson JR. Orexin/hypocretin and dysregulated eating: Promotion of foraging behavior. Brain Res 2018; 1731:145915. [PMID: 30125533 DOI: 10.1016/j.brainres.2018.08.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/31/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022]
Abstract
At its discovery, orexin/hypocretin (OX) was hypothesized to promote food intake. Subsequently, with the identification of the participation of OX in numerous other phenomena, including arousal and drug seeking, this neuropeptide was proposed to be involved in highly motivated behaviors. The present review develops the hypothesis that the primary evolutionary function of OX is to promote foraging behavior, seeking for food under conditions of limited availability. Thus, it will first describe published literature on OX and homeostatic food intake, which shows that OX neurons are activated by conditions of food deprivation and in turn stimulate food intake. Next, it will present literature on excessive and binge-like food intake, which demonstrates that OX stimulates both intake and willingness to work for palatable food. Importantly, studies show that binge-like eating can be inhibited by OX antagonists at doses far lower than those required to suppress homeostatic intake (3 mg/kg vs. 30 mg/kg), suggesting that an OX-based pharmacotherapy, at the right dose, could specifically control dysregulated eating. Finally, the review will discuss the role of OX in foraging behavior, citing literature which shows that OX neurons, which are activated during the anticipation of food reward, can promote a number of phenomena involved in successful foraging, including food-anticipatory locomotor behavior, olfactory sensitivity, visual attention, spatial memory, and mastication. Thus, OX may promote homeostatic eating, as well as binge eating of palatable food, due to its ability to stimulate and coordinate the activities involved in foraging behavior.
Collapse
Affiliation(s)
- Jessica R Barson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
26
|
Fuller-Jackson JP, Clarke IJ, Rao A, Henry BA. Exercise counteracts the homeostatic decrease in thermogenesis caused by caloric restriction in sheep. FASEB J 2018; 32:3859-3869. [PMID: 29455575 DOI: 10.1096/fj.201701504r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Caloric restriction causes a homeostatic reduction in thermogenesis. We aimed to determine whether exercise could counteract this. We studied four groups of normal-weight ewes ( n = 5), including control sedentary fed ad libitum, exercise fed ad libitum (30 min/d, 5 d/wk), diet-restricted (70% of ad libitum food intake), and combined diet and exercise. Temperature probes implanted in sternal and retroperitoneal adipose tissue and skeletal muscle measured thermogenesis. After the 4-wk intervention, hypothalami were collected for in situ hybridization, and fat and muscle biopsies were collected for real-time PCR and Western blotting. Combined diet and exercise reduced adiposity ( P < 0.05). Caloric restriction alone reduced overnight temperatures in sternal and retroperitoneal fat ( P < 0.05), which was counteracted by exercise ( P < 0.05). Exercise did not induce expression of cellular markers of browning in adipose tissue. There was no effect of diet or exercise on skeletal muscle thermogenesis. Combined diet and exercise increased the expression of neuropeptide Y and agouti-related protein in the hypothalamic arcuate nucleus ( P < 0.05), consistent with reduced adiposity. Gene expressions of key hypothalamic appetite-regulating peptides were not associated with altered thermogenesis. We demonstrate that exercise counteracts the inhibitory effect of caloric restriction to restore thermogenesis in adipose tissue of sheep.-Fuller-Jackson, J.-P., Clarke, I. J., Rao, A., Henry, B. A. Exercise counteracts the homeostatic decrease in thermogenesis caused by caloric restriction in sheep.
Collapse
Affiliation(s)
- John-Paul Fuller-Jackson
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Iain J Clarke
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Alexandra Rao
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Belinda A Henry
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
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]
|
28
|
Abstract
PURPOSE OF REVIEW Spontaneous physical activity (SPA) is a physical activity not motivated by a rewarding goal, such as that associated with food-seeking or wheel-running behavior. SPA is often thought of as only "fidgeting," but that is a mischaracterization, since fidgety behavior can be linked to stereotypies in neurodegenerative disease and other movement disorders. Instead, SPA should be thought of as all physical activity behavior that emanates from an unconscious drive for movement. RECENT FINDINGS An example of this may be restless behavior, which can include fidgeting and gesticulating, frequent sit-to-stand movement, and more time spent standing and moving. All physical activity burns calories, and as such, SPA could be manipulated as a means to burn calories, and defend against weight gain and reduce excess adiposity. In this review, we discuss human and animal literature on the use of SPA in reducing weight gain, the neuromodulators that could be targeted to this end, and future directions in this field.
Collapse
Affiliation(s)
- Catherine M Kotz
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA.
- GRECC, Minneapolis VA Health Care System, GRECC, One Veterans Drive, Minneapolis, MN, 55417, USA.
| | | | - Jennifer A Teske
- Department of Nutritional Sciences, University of Arizona, 1177 E 4th street, Shantz 332, Tucson, AZ, 85721, USA
| | - Charles J Billington
- Department of Medicine, University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 5545, USA
- Minneapolis VA Health Care System, One Veterans Drive, Minneapolis, MN, 55417, USA
| |
Collapse
|
29
|
Goforth PB, Myers MG. Roles for Orexin/Hypocretin in the Control of Energy Balance and Metabolism. Curr Top Behav Neurosci 2017; 33:137-156. [PMID: 27909992 DOI: 10.1007/7854_2016_51] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The neuropeptide hypocretin is also commonly referred to as orexin, since its orexigenic action was recognized early. Orexin/hypocretin (OX) neurons project widely throughout the brain and the physiologic and behavioral functions of OX are much more complex than initially conceived based upon the stimulation of feeding. OX most notably controls functions relevant to attention, alertness, and motivation. OX also plays multiple crucial roles in the control of food intake, metabolism, and overall energy balance in mammals. OX signaling not only promotes food-seeking behavior upon short-term fasting to increase food intake and defend body weight, but, conversely, OX signaling also supports energy expenditure to protect against obesity. Furthermore, OX modulates the autonomic nervous system to control glucose metabolism, including during the response to hypoglycemia. Consistently, a variety of nutritional cues (including the hormones leptin and ghrelin) and metabolites (e.g., glucose, amino acids) control OX neurons. In this chapter, we review the control of OX neurons by nutritional/metabolic cues, along with our current understanding of the mechanisms by which OX and OX neurons contribute to the control of energy balance and metabolism.
Collapse
Affiliation(s)
- Paulette B Goforth
- Department of Pharmacology, University of Michigan, 1000 Wall St, 5131 Brehm Tower, Ann Arbor, MI, 48105, USA
| | - Martin G Myers
- Departments of Internal Medicine, and Molecular and Integrative Physiology, University of Michigan, 1000 Wall St, 6317 Brehm Tower, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
30
|
Bunney PE, Zink AN, Holm AA, Billington CJ, Kotz CM. Orexin activation counteracts decreases in nonexercise activity thermogenesis (NEAT) caused by high-fat diet. Physiol Behav 2017; 176:139-148. [PMID: 28363838 PMCID: PMC5510739 DOI: 10.1016/j.physbeh.2017.03.040] [Citation(s) in RCA: 848] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/27/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Overweight and obesity result from an imbalance between caloric intake and energy expenditure, including expenditure from spontaneous physical activity (SPA). Changes in SPA and resulting changes in non-exercise activity thermogenesis (NEAT) likely interact with diet to influence risk for obesity. However, previous research on the relationship between diet, physical activity, and energy expenditure has been mixed. The neuropeptide orexin is a driver of SPA, and orexin neuron activity can be manipulated using DREADDs (Designer Receptors Exclusively Activated by Designer Drugs). We hypothesized that HFD decreases SPA and NEAT, and that DREADD-mediated activation of orexin neuron signaling would abolish this decrease and produce an increase in NEAT instead. To test these ideas, we characterized behaviors to determine the extent to which access to a high-fat diet (HFD) influences the proportion and probability of engaging in food intake and activity. We then measured NEAT following access to HFD and following a DREADD intervention targeting orexin neurons. Two cohorts of orexin-cre male mice were injected with an excitatory DREADD virus into the caudal hypothalamus, where orexin neurons are concentrated. Mice were then housed in continuous metabolic phenotyping cages (Sable Promethion). Food intake, indirect calorimetry, and SPA were automatically measured every second. For cohort 1 (n=8), animals were given access to chow, then switched to HFD. For cohort 2 (n=4/group), half of the animals were given access to HFD, the other access to chow. Then, among animals on HFD, orexin neurons were activated following injections of clozapine n-oxide (CNO). Mice on HFD spent significantly less time eating (p<0.01) and more time inactive compared to mice on chow (p<0.01). Following a meal, mice on HFD were significantly more likely to engage in periods of inactivity compared to those on chow (p<0.05). NEAT was decreased in animals on HFD, and was increased to the NEAT level of control animals following activation of orexin neurons with DREADDs. Food intake (kilocalories) was not significantly different between mice on chow and HFD, yet mice on chow expended more energy per unit of SPA, relative to that in mice consuming HFD. These results suggest that HFD consumption reduces SPA and NEAT, and increases inactivity following a meal. Together, the data suggest a change in the efficiency of energy expenditure based upon diet, such that SPA during HFD burns fewer calories compared to SPA on a standard chow diet.
Collapse
Affiliation(s)
- P E Bunney
- Minnesota Obesity Prevention Training Program, University of Minnesota, United States; Geriatric Research and Clinical Center, Minneapolis VA Health Care System, United States.
| | - A N Zink
- Medical Devices Center, University of Minnesota, United States
| | - A A Holm
- Food Science and Nutrition, University of Minnesota, United States
| | - C J Billington
- Department of Medicine, University of Minnesota, United States; Minnesota Obesity Center, University of Minnesota, United States; Department of Medicine, Minneapolis VA Health Care System, United States
| | - C M Kotz
- Geriatric Research and Clinical Center, Minneapolis VA Health Care System, United States; Minnesota Obesity Center, University of Minnesota, United States; Department of Integrative Biology and Physiology, University of Minnesota, United States
| |
Collapse
|
31
|
Coborn JE, DePorter DP, Mavanji V, Sinton CM, Kotz CM, Billington CJ, Teske JA. Role of orexin-A in the ventrolateral preoptic area on components of total energy expenditure. Int J Obes (Lond) 2017; 41:1256-1262. [PMID: 28392556 DOI: 10.1038/ijo.2017.92] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 03/20/2017] [Accepted: 03/26/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Identifying whether components of total energy expenditure (EE) are affected by orexin receptor (OXR1 and OXR2) stimulation or antagonism with dual orexin receptor antagonists (DORAs) has relevance for obesity treatment. Orexin receptor stimulation reduces weight gain by increasing total EE and EE during spontaneous physical activity (SPA). OBJECTIVE The purpose of this study was to determine if a DORA (TCS-1102) in the ventrolateral preoptic area (VLPO) reduced orexin-A-induced arousal, SPA, total EE and EE during sleep, rest, wake and SPA and whether the DORA alone reduced total EE and its components. We hypothesized that: (1) a DORA would reduce orexin-A induced increases in arousal, SPA, components of total EE, reductions in sleep and the EE during sleep and (2) the DORA alone would reduce baseline (non-stimulated) SPA and total EE. SUBJECTS/METHODS Sleep, wakefulness, SPA and EE were determined after microinjection of the DORA (TCS-1102) and orexin-A in the VLPO of male Sprague-Dawley rats with a unilateral cannula targeted towards the VLPO. Individual components of total EE were determined based on time-stamped data. RESULTS The DORA reduced orexin-A-induced increases in arousal, SPA, total EE and EE during SPA, wake, rest and sleep 1 h post injection (P<0.05). Orexin-A significantly reduced sleep and significantly increased EE during sleep 1 h post injection (P<0.05). Furthermore, the DORA alone significantly reduced total EE, EE during sleep (NREM and REM) and resting EE 2 h post injection (P<0.05). CONCLUSIONS These data suggest that orexin-A reduces weight gain by stimulating total EE through increases in EE during SPA, rest and sleep. Residual effects of the DORA alone include decreases in total EE and EE during sleep and rest, which may promote weight gain.
Collapse
Affiliation(s)
- J E Coborn
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - D P DePorter
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - V Mavanji
- Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - C M Sinton
- Arizona Respiratory Center, University of Arizona, Tucson, AZ, USA
| | - C M Kotz
- Minneapolis VA Health Care System, Minneapolis, MN, USA.,Geriatric Research Education and Clinical Center, Minneapolis, MN, USA.,Minnesota Obesity Center, Saint Paul, MN, USA.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - C J Billington
- Minneapolis VA Health Care System, Minneapolis, MN, USA.,Minnesota Obesity Center, Saint Paul, MN, USA.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA.,Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - J A Teske
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA.,Minneapolis VA Health Care System, Minneapolis, MN, USA.,Minnesota Obesity Center, Saint Paul, MN, USA.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| |
Collapse
|
32
|
Elbaz I, Levitas-Djerbi T, Appelbaum L. The Hypocretin/Orexin Neuronal Networks in Zebrafish. Curr Top Behav Neurosci 2017; 33:75-92. [PMID: 28012092 DOI: 10.1007/7854_2016_59] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The hypothalamic Hypocretin/Orexin (Hcrt) neurons secrete two Hcrt neuropeptides. These neurons and peptides play a major role in the regulation of feeding, sleep wake cycle, reward-seeking, addiction, and stress. Loss of Hcrt neurons causes the sleep disorder narcolepsy. The zebrafish has become an attractive model to study the Hcrt neuronal network because it is a transparent vertebrate that enables simple genetic manipulation, imaging of the structure and function of neuronal circuits in live animals, and high-throughput monitoring of behavioral performance during both day and night. The zebrafish Hcrt network comprises ~16-60 neurons, which similar to mammals, are located in the hypothalamus and widely innervate the brain and spinal cord, and regulate various fundamental behaviors such as feeding, sleep, and wakefulness. Here we review how the zebrafish contributes to the study of the Hcrt neuronal system molecularly, anatomically, physiologically, and pathologically.
Collapse
Affiliation(s)
- Idan Elbaz
- The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Talia Levitas-Djerbi
- The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Lior Appelbaum
- The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| |
Collapse
|
33
|
Miyatake Y, Shiuchi T, Mawatari K, Toda S, Taniguchi Y, Futami A, Sato F, Kuroda M, Sebe M, Tsutsumi R, Harada N, Minokoshi Y, Kitamura T, Gotoh K, Ueno M, Nakaya Y, Sakaue H. Intracerebroventricular injection of ghrelin decreases wheel running activity in rats. Peptides 2017; 87:12-19. [PMID: 27825986 DOI: 10.1016/j.peptides.2016.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/28/2016] [Accepted: 11/01/2016] [Indexed: 01/15/2023]
Abstract
There is an increasing interest in elucidating the molecular mechanisms by which voluntary exercise is regulated. In this study, we examined how the central nervous system regulates exercise. We used SPORTS rats, which were established in our laboratory as a highly voluntary murine exercise model. SPORTS rats showed lower levels of serum ghrelin compared with those of the parental line of Wistar rats. Intracerebroventricular and intraperitoneal injection of ghrelin decreased wheel-running activity in SPORTS rats. In addition, daily injection of the ghrelin inhibitor JMV3002 into the lateral ventricles of Wistar rats increased wheel-running activity. Co-administration of obestatin inhibited ghrelin-induced increases in food intake but did not inhibit ghrelin-induced suppression of voluntary exercise in rats. Growth hormone secretagogue receptor (GHSR) in the hypothalamus and hippocampus of SPORTS rats was not difference that in control rats. We created an arcuate nucleus destruction model by administering monosodium glutamate (MSG) to neonatal SPORTS rats. Injection of ghrelin into MSG-treated rats decreased voluntary exercise but did not increase food intake, suggesting that wheel-running activity is not controlled by the arcuate nucleus neurons that regulate feeding. These results provide new insights into the mechanism by which ghrelin regulates voluntary activity independent of arcuate nucleus neurons.
Collapse
Affiliation(s)
- Yumiko Miyatake
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan; PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, 332-0012, Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Satomi Toda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Yasuko Taniguchi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Akari Futami
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Fukiko Sato
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Mayu Sebe
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Nagakatsu Harada
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Yasuhiko Minokoshi
- Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8585, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Maebashi, Gunma University, 371-8512, Japan
| | - Koro Gotoh
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Oita, 879-5593, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Miki, Kagawa, 761-0793, Japan
| | - Yutaka Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima, 770-8503, Japan.
| |
Collapse
|
34
|
Perez-Leighton C, Little MR, Grace M, Billington C, Kotz CM. Orexin signaling in rostral lateral hypothalamus and nucleus accumbens shell in the control of spontaneous physical activity in high- and low-activity rats. Am J Physiol Regul Integr Comp Physiol 2016; 312:R338-R346. [PMID: 28039192 DOI: 10.1152/ajpregu.00339.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 01/04/2023]
Abstract
Spontaneous physical activity (SPA) describes activity outside of formal exercise and shows large interindividual variability. The hypothalamic orexin/hypocretin peptides are key regulators of SPA. Orexins drive SPA within multiple brain sites, including rostral lateral hypothalamus (LH) and nucleus accumbens shell (NAcSh). Rats with high basal SPA (high activity, HA) show higher orexin mRNA expression and SPA after injection of orexin-A in rostral LH compared with low-activity (LA) rats. Here, we explored the contribution of orexin signaling in rostral LH and NAcSh to the HA/LA phenotype. We found that HA rats have higher sensitivity to SPA after injection of orexin-A in rostral LH, but not in NAcSh. HA and LA rats showed similar levels of orexin receptor expression in rostral LH, and activation of orexin-producing neurons after orexin-A injection in rostral LH. Also, in HA and LA rats, the coinjection of orexin-A in rostral LH and NAcSh failed to further increase SPA beyond the effects of orexin-A in rostral LH. Pretreatment with muscimol, a GABAA receptor agonist, in NAcSh potentiated SPA produced by orexin-A injection in rostral LH in HA but not in LA rats. Our results suggest that a feedback loop from orexin-responsive neurons in rostral LH to orexin neurons and a the NAcSh-orexin neuron-rostral LH circuit regulate SPA. Overall, our data suggest that differences in orexin sensitivity in rostral LH and its modulation by GABA afferents from NAcSh contribute to individual SPA differences.
Collapse
Affiliation(s)
- Claudio Perez-Leighton
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andres Bello, Santiago, Region Metropolitana, Chile.,Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota; and
| | - Morgan R Little
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota.,Minnesota Obesity Center, University of Minnesota, Minnesota
| | - Martha Grace
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota
| | - Charles Billington
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota.,Minnesota Obesity Center, University of Minnesota, Minnesota.,Department of Medicine, University of Minnesota, Minneapolis, St. Paul, Minnesota
| | - Catherine M Kotz
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota; .,Minnesota Obesity Center, University of Minnesota, Minnesota.,Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
35
|
Pan L, Qi R, Wang J, Zhou W, Liu J, Cai Y. Evidence for a Role of Orexin/Hypocretin System in Vestibular Lesion-Induced Locomotor Abnormalities in Rats. Front Neurosci 2016; 10:355. [PMID: 27507932 PMCID: PMC4960243 DOI: 10.3389/fnins.2016.00355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/13/2016] [Indexed: 12/21/2022] Open
Abstract
Vestibular damage can induce locomotor abnormalities in both animals and humans. Rodents with bilateral vestibular loss showed vestibular deficits syndrome such as circling, opisthotonus as well as locomotor and exploratory hyperactivity. Previous studies have investigated the changes in the dopamine system after vestibular loss, but the results are inconsistent and inconclusive. Numerous evidences indicate that the orexin system is implicated in central motor control. We hypothesized that orexin may be potentially involved in vestibular loss-induced motor disorders. In this study, we examined the effects of arsanilate- or 3,3′-iminodipropionitrile (IDPN)-induced vestibular lesion (AVL or IVL) on the orexin-A (OXA) labeling in rat hypothalamus using immunohistochemistry. The vestibular lesion-induced locomotor abnormalities were recorded and verified using a histamine H4 receptor antagonist JNJ7777120 (20 mg/kg, i.p.). The effects of the orexin receptor type 1 antagonist SB334867 (16 μg, i.c.v.) on these behavior responses were also investigated. At 72 h post-AVL and IVL, animals exhibited vestibular deficit syndrome and locomotor hyperactivity in the home cages. These responses were significantly alleviated by JNJ7777120 which also eliminated AVL-induced increases in exploratory behavior in an open field. The numbers of OXA-labeled neurons in the hypothalamus were significantly increased in the AVL animals at 72 h post-AVL and in the IVL animals at 24, 48, and 72 h post-IVL. SB334867 significantly attenuated the vestibular deficit syndrome and locomotor hyperactivity at 72 h post-AVL and IVL. It also decreased exploratory behavior in the AVL animals. These results suggested that the alteration of OXA expression might contribute to locomotor abnormalities after acute vestibular lesion. The orexin receptors might be the potential therapeutic targets for vestibular disorders.
Collapse
Affiliation(s)
- Leilei Pan
- Department of Nautical Injury Prevention, Faculty of Navy Medicine, Second Military Medical University Shanghai, China
| | - Ruirui Qi
- Department of Nautical Injury Prevention, Faculty of Navy Medicine, Second Military Medical University Shanghai, China
| | - Junqin Wang
- Department of Nautical Injury Prevention, Faculty of Navy Medicine, Second Military Medical University Shanghai, China
| | - Wei Zhou
- Department of Nautical Injury Prevention, Faculty of Navy Medicine, Second Military Medical University Shanghai, China
| | - Jiluo Liu
- Department of Nautical Injury Prevention, Faculty of Navy Medicine, Second Military Medical University Shanghai, China
| | - Yiling Cai
- Department of Nautical Injury Prevention, Faculty of Navy Medicine, Second Military Medical University Shanghai, China
| |
Collapse
|
36
|
Gac L, Butterick TA, Duffy CM, Teske JA, Perez-Leighton CE. Role of the non-opioid dynorphin peptide des-Tyr-dynorphin (DYN-A(2-17)) in food intake and physical activity, and its interaction with orexin-A. Peptides 2016; 76:14-8. [PMID: 26654796 DOI: 10.1016/j.peptides.2015.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023]
Abstract
Food intake and physical activity are regulated by multiple neuropeptides, including orexin and dynorphin (DYN). Orexin-A (OXA) is one of two orexin peptides with robust roles in regulation of food intake and spontaneous physical activity (SPA). DYN collectively refers to several peptides, some of which act through opioid receptors (opioid DYN) and some whose biological effects are not mediated by opioid receptors (non-opioid DYN). While opioid DYN is known to increase food intake, the effects of non-opioid DYN peptides on food intake and SPA are unknown. Neurons that co-express and release OXA and DYN are located within the lateral hypothalamus. Limited evidence suggests that OXA and opioid DYN peptides can interact to modulate some aspects of behaviors classically related to orexin peptide function. The paraventricular hypothalamic nucleus (PVN) is a brain area where OXA and DYN peptides might interact to modulate food intake and SPA. We demonstrate that injection of des-Tyr-dynorphin (DYN-A(2-17), a non opioid DYN peptide) into the PVN increases food intake and SPA in adult mice. Co-injection of DYN-A(2-17) and OXA in the PVN further increases food intake compared to DYN-A(2-17) or OXA alone. This is the first report describing the effects of non-opioid DYN-A(2-17) on food intake and SPA, and suggests that DYN-A(2-17) interacts with OXA in the PVN to modulate food intake. Our data suggest a novel function for non-opioid DYN-A(2-17) on food intake, supporting the concept that some behavioral effects of the orexin neurons result from combined actions of the orexin and DYN peptides.
Collapse
Affiliation(s)
- L Gac
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andres Bello, Santiago, Region Metropolitana, Chile
| | - T A Butterick
- Minneapolis VA Health Care System, Minneapolis, MN, USA; Minnesota Obesity Center, University of Minnesota, Saint Paul, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - C M Duffy
- Minneapolis VA Health Care System, Minneapolis, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - J A Teske
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA; Minneapolis VA Health Care System, Minneapolis, MN, USA; Minnesota Obesity Center, University of Minnesota, Saint Paul, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - C E Perez-Leighton
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andres Bello, Santiago, Region Metropolitana, Chile; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA.
| |
Collapse
|
37
|
Yoshimura M, Uezono Y, Ueta Y. Anorexia in human and experimental animal models: physiological aspects related to neuropeptides. J Physiol Sci 2015; 65:385-95. [PMID: 26123258 PMCID: PMC10717229 DOI: 10.1007/s12576-015-0386-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/16/2015] [Indexed: 01/20/2023]
Abstract
Anorexia, a loss of appetite for food, can be caused by various physiological and pathophysiological conditions. In this review, firstly, clinical aspects of anorexia nervosa are summarized in brief. Secondly, hypothalamic neuropeptides responsible for feeding regulation in each hypothalamic nucleus are discussed. Finally, three different types of anorexigenic animal models; dehydration-induced anorexia, cisplatin-induced anorexia and cancer anorexia-cachexia, are introduced. In conclusion, hypothalamic neuropeptides may give us novel insight to understand and find effective therapeutics strategy essential for various kinds of anorexia.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555 Japan
| | - Yasuhito Uezono
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, 104-0045 Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555 Japan
| |
Collapse
|
38
|
Mavanji V, Perez-Leighton CE, Kotz CM, Billington CJ, Parthasarathy S, Sinton CM, Teske JA. Promotion of Wakefulness and Energy Expenditure by Orexin-A in the Ventrolateral Preoptic Area. Sleep 2015; 38:1361-70. [PMID: 25845696 DOI: 10.5665/sleep.4970] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 03/07/2015] [Indexed: 12/25/2022] Open
Abstract
STUDY OBJECTIVES The ventrolateral preoptic area (VLPO) and the orexin/hypocretin neuronal system are key regulators of sleep onset, transitions between vigilance states, and energy homeostasis. Reciprocal projections exist between the VLPO and orexin/hypocretin neurons. Although the importance of the VLPO to sleep regulation is clear, it is unknown whether VLPO neurons are involved in energy balance. The purpose of these studies was to determine if the VLPO is a site of action for orexin-A, and which orexin receptor subtype(s) would mediate these effects of orexin-A. We hypothesized that orexin-A in the VLPO modulates behaviors (sleep and wakefulness, feeding, spontaneous physical activity [SPA]) to increase energy expenditure. DESIGN AND MEASUREMENTS Sleep, wakefulness, SPA, feeding, and energy expenditure were determined after orexin-A microinjection in the VLPO of male Sprague-Dawley rats with unilateral cannulae targeting the VLPO. We also tested whether pretreatment with a dual orexin receptor antagonist (DORA, TCS-1102) or an OX2R antagonist (JNJ-10397049) blocked the effects of orexin-A on the sleep/wake cycle or SPA, respectively. RESULTS Orexin-A injected into the VLPO significantly increased wakefulness, SPA, and energy expenditure (SPA-induced and total) and reduced NREM sleep and REM sleep with no effect on food intake. Pretreatment with DORA blocked the increase in wakefulness and the reduction in NREM sleep elicited by orexin-A, and the OX2R antagonist reduced SPA stimulated by orexin-A. CONCLUSIONS These data show the ventrolateral preoptic area is a site of action for orexin-A, which may promote negative energy balance by modulating sleep/wakefulness and stimulating spontaneous physical activity and energy expenditure.
Collapse
Affiliation(s)
| | - Claudio E Perez-Leighton
- Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile.,Escuela de Nutricion, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Catherine M Kotz
- Minneapolis VA Health Care System, Minneapolis, MN.,Geriatric Research Education and Clinical Center, Minneapolis, MN.,Minnesota Obesity Center, University of Minnesota, Saint Paul, MN.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN
| | - Charles J Billington
- Minneapolis VA Health Care System, Minneapolis, MN.,Minnesota Obesity Center, University of Minnesota, Saint Paul, MN.,Department of Medicine, University of Minnesota, Saint Paul, MN.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN
| | - Sairam Parthasarathy
- Arizona Respiratory Center University of Arizona, Tucson, AZ.,Department of Medicine University of Arizona, Tucson, AZ
| | - Christopher M Sinton
- Arizona Respiratory Center University of Arizona, Tucson, AZ.,Department of Medicine University of Arizona, Tucson, AZ
| | - Jennifer A Teske
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ.,Minneapolis VA Health Care System, Minneapolis, MN.,Minnesota Obesity Center, University of Minnesota, Saint Paul, MN.,Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN
| |
Collapse
|
39
|
Abstract
Sitting too much kills. Epidemiological, physiological and molecular data suggest that sedentary lifestyle can explain, in part, how modernity is associated with obesity, more than 30 chronic diseases and conditions and high healthcare costs. Excessive sitting--sitting disease--is not innate to the human condition. People were designed to be bipedal and, before the industrial revolution, people moved substantially more throughout the day than they do presently. It is encouraging that solutions exist to reverse sitting disease. Work environments, schools, communities and cities can be re-imagined and re-invented as walking spaces, and people thereby offered more active, happier, healthier and more productive lives.
Collapse
Affiliation(s)
- James A Levine
- Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA,
| |
Collapse
|
40
|
Villablanca PA, Alegria JR, Mookadam F, Holmes DR, Wright RS, Levine JA. Nonexercise activity thermogenesis in obesity management. Mayo Clin Proc 2015; 90:509-19. [PMID: 25841254 DOI: 10.1016/j.mayocp.2015.02.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
Obesity is linked to cardiovascular disease. The global increase in sedentary lifestyle is an important factor contributing to the rising prevalence of the obesity epidemic. Traditionally, counseling has focused on moderate- to vigorous-intensity exercise, with disappointing results. Nonexercise activity thermogenesis (NEAT) is an important component of daily energy expenditure. It represents the common daily activities, such as fidgeting, walking, and standing. These high-effect NEAT movements could result in up to an extra 2000 kcal of expenditure per day beyond the basal metabolic rate, depending on body weight and level of activity. Implementing NEAT during leisure-time and occupational activities could be essential to maintaining a negative energy balance. NEAT can be applied by being upright, ambulating, and redesigning workplace and leisure-time environments to promote NEAT. The benefits of NEAT include not only the extra calories expended but also the reduced occurrence of the metabolic syndrome, cardiovascular events, and all-cause mortality. We believe that to overcome the obesity epidemic and its adverse cardiovascular consequences, NEAT should be part of the current medical recommendations. The content of this review is based on a literature search of PubMed and the Google search engine between January 1, 1960, and October 1, 2014, using the search terms physical activity, obesity, energy expenditure, nonexercise activity thermogenesis, and NEAT.
Collapse
Affiliation(s)
- Pedro A Villablanca
- Division of Cardiovascular Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, NY.
| | | | - Farouk Mookadam
- Division of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ
| | - David R Holmes
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - R Scott Wright
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
41
|
Brown JA, Woodworth HL, Leinninger GM. To ingest or rest? Specialized roles of lateral hypothalamic area neurons in coordinating energy balance. Front Syst Neurosci 2015; 9:9. [PMID: 25741247 PMCID: PMC4332303 DOI: 10.3389/fnsys.2015.00009] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/15/2015] [Indexed: 12/26/2022] Open
Abstract
Survival depends on an organism’s ability to sense nutrient status and accordingly regulate intake and energy expenditure behaviors. Uncoupling of energy sensing and behavior, however, underlies energy balance disorders such as anorexia or obesity. The hypothalamus regulates energy balance, and in particular the lateral hypothalamic area (LHA) is poised to coordinate peripheral cues of energy status and behaviors that impact weight, such as drinking, locomotor behavior, arousal/sleep and autonomic output. There are several populations of LHA neurons that are defined by their neuropeptide content and contribute to energy balance. LHA neurons that express the neuropeptides melanin-concentrating hormone (MCH) or orexins/hypocretins (OX) are best characterized and these neurons play important roles in regulating ingestion, arousal, locomotor behavior and autonomic function via distinct neuronal circuits. Recently, another population of LHA neurons containing the neuropeptide Neurotensin (Nts) has been implicated in coordinating anorectic stimuli and behavior to regulate hydration and energy balance. Understanding the specific roles of MCH, OX and Nts neurons in harmonizing energy sensing and behavior thus has the potential to inform pharmacological strategies to modify behaviors and treat energy balance disorders.
Collapse
Affiliation(s)
- Juliette A Brown
- Department of Pharmacology and Toxicology, Michigan State University East Lansing, MI, USA ; Center for Integrative Toxicology East Lansing, MI, USA
| | | | - Gina M Leinninger
- Center for Integrative Toxicology East Lansing, MI, USA ; Department of Physiology, Michigan State University East Lansing, MI, USA
| |
Collapse
|
42
|
Miyatake Y, Shiuchi T, Ueta T, Taniguchi Y, Futami A, Sato F, Kitamura T, Tsutsumi R, Harada N, Nakaya Y, Sakaue H. Intracerebroventricular injection of adiponectin regulates locomotor activity in rats. THE JOURNAL OF MEDICAL INVESTIGATION 2015; 62:199-203. [DOI: 10.2152/jmi.62.199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Yumiko Miyatake
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School
- PRESTO, Japan Science and Technology Agency
| | - Tomoyo Ueta
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Yasuko Taniguchi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Akari Futami
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Fukiko Sato
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Nagakatsu Harada
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Yutaka Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| |
Collapse
|
43
|
Apolzan JW, Bray GA, Smith SR, de Jonge L, Rood J, Han H, Redman LM, Martin CK. Effects of weight gain induced by controlled overfeeding on physical activity. Am J Physiol Endocrinol Metab 2014; 307:E1030-7. [PMID: 25294214 PMCID: PMC4254990 DOI: 10.1152/ajpendo.00386.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is unclear whether physical activity changes following long-term overfeeding and in response to different dietary protein intakes. Twenty-five (16 males, 9 females) healthy adults (18-35 yr) with BMI ranging from 19 to 30 kg/m(2) enrolled in this inpatient study. In a parallel group design, participants were fed 140% of energy needs, with 5, 15, or 25% of energy from protein, for 56 days. Participants wore an RT3 accelerometer for at least 59 days throughout baseline and during overfeeding and completed 24-h whole room metabolic chamber assessments at baseline and on days 1, 14, and 56 of overfeeding and on day 57, when the baseline energy intake was consumed, to measure percent of time active and spontaneous physical activity (SPA; kcal/day). Changes in activity were also assessed by doubly labeled water (DLW). From accelerometry, vector magnitude (VM), a weight-independent measure of activity, and activity energy expenditure (AEE) increased with weight gain during overfeeding. AEE remained increased after adjusting for changes in body composition. Activity-related energy expenditure (AREE) from DLW and percent activity and SPA in the metabolic chamber increased with overfeeding, but SPA was no longer significant after adjusting for change in body composition. Change in VM and AEE were positively correlated with weight gain; however, change in activity was not affected by protein intake. Overfeeding produces an increase in physical activity and in energy expended in physical activity after adjusting for changes in body composition, suggesting that increased activity in response to weight gain might be one mechanism to support adaptive thermogenesis.
Collapse
Affiliation(s)
- John W Apolzan
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - George A Bray
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Steven R Smith
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Lilian de Jonge
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Jennifer Rood
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Hongmei Han
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Leanne M Redman
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Corby K Martin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| |
Collapse
|
44
|
Zink AN, Perez-Leighton CE, Kotz CM. The orexin neuropeptide system: physical activity and hypothalamic function throughout the aging process. Front Syst Neurosci 2014; 8:211. [PMID: 25408639 PMCID: PMC4219460 DOI: 10.3389/fnsys.2014.00211] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022] Open
Abstract
There is a rising medical need for novel therapeutic targets of physical activity. Physical activity spans from spontaneous, low intensity movements to voluntary, high-intensity exercise. Regulation of spontaneous and voluntary movement is distributed over many brain areas and neural substrates, but the specific cellular and molecular mechanisms responsible for mediating overall activity levels are not well understood. The hypothalamus plays a central role in the control of physical activity, which is executed through coordination of multiple signaling systems, including the orexin neuropeptides. Orexin producing neurons integrate physiological and metabolic information to coordinate multiple behavioral states and modulate physical activity in response to the environment. This review is organized around three questions: (1) How do orexin peptides modulate physical activity? (2) What are the effects of aging and lifestyle choices on physical activity? (3) What are the effects of aging on hypothalamic function and the orexin peptides? Discussion of these questions will provide a summary of the current state of knowledge regarding hypothalamic orexin regulation of physical activity during aging and provide a platform on which to develop improved clinical outcomes in age-associated obesity and metabolic syndromes.
Collapse
Affiliation(s)
- Anastasia N Zink
- Graduate Program in Neuroscience, School of Medicine, University of Minnesota Minneapolis, MN, USA
| | | | - Catherine M Kotz
- Graduate Program in Neuroscience, School of Medicine, University of Minnesota Minneapolis, MN, USA ; GRECC (11G), Minneapolis VA Healthcare System Minneapolis, MN, USA ; Department of Food Science and Nutrition, University of Minnesota Saint Paul, MN, USA
| |
Collapse
|
45
|
Chen YW, Barson JR, Chen A, Hoebel BG, Leibowitz SF. Hypothalamic peptides controlling alcohol intake: differential effects on microstructure of drinking bouts. Alcohol 2014; 48:657-64. [PMID: 25241055 DOI: 10.1016/j.alcohol.2014.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Different alcohol drinking patterns, involving either small and frequent drinking bouts or large and long-lasting bouts, are found to differentially affect the risk for developing alcohol-related diseases, suggesting that they have different underlying mechanisms. Such mechanisms may involve orexigenic peptides known to stimulate alcohol intake through their actions in the hypothalamic paraventricular nucleus (PVN). These include orexin (OX), which is expressed in the perifornical lateral hypothalamus, and galanin (GAL) and enkephalin (ENK), which are expressed within as well as outside the PVN. To investigate the possibility that these peptides affect different aspects of consumption, a microstructural analysis of ethanol drinking behavior was performed in male, Sprague-Dawley rats trained to drink 7% ethanol and implanted with guide shafts aimed at the PVN. While housed in specialized cages containing computerized intake monitors (BioDAQ Laboratory Intake Monitoring System, Research Diets Inc., New Brunswick, NJ) that measure bouts of ethanol drinking, these rats were given PVN injections of OX (0.9 nmol), GAL (1.0 nmol), or the ENK analog D-Ala2-met-enkephalinamide (DALA) (14.2 nmol), as compared to saline vehicle. Results revealed clear differences between the effects of these peptides. While all 3 stimulated ethanol intake, they had distinct effects on patterns of drinking, with OX increasing the number of drinking bouts, GAL increasing the size of the drinking bouts, and DALA increasing both the size and duration of the bouts. In contrast, these peptides had little impact on water or food intake. These results support the idea that different peptides can increase ethanol consumption by promoting distinct aspects of the ethanol drinking response. The stimulatory effect of OX on drinking frequency may be related to its neuronally stimulatory properties, while the stimulatory effect of GAL and ENK on bout size and duration may reflect a suppressive effect of these neuronally inhibitory peptides on the satiety-controlling PVN.
Collapse
|
46
|
Jalewa J, Wong-Lin K, McGinnity TM, Prasad G, Hölscher C. Increased number of orexin/hypocretin neurons with high and prolonged external stress-induced depression. Behav Brain Res 2014; 272:196-204. [DOI: 10.1016/j.bbr.2014.05.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 01/14/2023]
|
47
|
Li J, Hu Z, de Lecea L. The hypocretins/orexins: integrators of multiple physiological functions. Br J Pharmacol 2014; 171:332-50. [PMID: 24102345 DOI: 10.1111/bph.12415] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 07/16/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022] Open
Abstract
The hypocretins (Hcrts), also known as orexins, are two peptides derived from a single precursor produced in the posterior lateral hypothalamus. Over the past decade, the orexin system has been associated with numerous physiological functions, including sleep/arousal, energy homeostasis, endocrine, visceral functions and pathological states, such as narcolepsy and drug abuse. Here, we review the discovery of Hcrt/orexins and their receptors and propose a hypothesis as to how the orexin system orchestrates these multifaceted physiological functions.
Collapse
Affiliation(s)
- Jingcheng Li
- Department of Physiology, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
48
|
Abstract
The basic elements of animal behavior that are critical to survival include energy, arousal, and motivation: Energy intake and expenditure are fundamental to all organisms for the performance of any type of function; according to the Yerkes-Dodson law, an optimal level of arousal is required for animals to perform normal functions; and motivation is critical to goal-oriented behaviors in higher animals. The brain is the primary organ that controls these elements and, through evolution, has developed specialized structures to accomplish this task. The orexin/hypocretin system in the perifornical/lateral hypothalamus, which was discovered 15 years ago, is one such specialized area. This review summarizes a fast-growing body of evidence discerning how the orexin/hypocretin system integrates internal and external cues to regulate energy intake that can then be used to generate sufficient arousal for animals to perform innate and goal-oriented behaviors.
Collapse
Affiliation(s)
- Xiao-Bing Gao
- Yale Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; ,
| | | |
Collapse
|
49
|
Weymann KB, Wood LJ, Zhu X, Marks DL. A role for orexin in cytotoxic chemotherapy-induced fatigue. Brain Behav Immun 2014; 37:84-94. [PMID: 24216337 PMCID: PMC3951615 DOI: 10.1016/j.bbi.2013.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/16/2013] [Accepted: 11/01/2013] [Indexed: 02/08/2023] Open
Abstract
Fatigue is the most common symptom related to cytotoxic chemotherapeutic treatment of cancer. Peripheral inflammation associated with cytotoxic chemotherapy is likely a causal factor of fatigue. The neural mechanisms by which cytotoxic chemotherapy associated inflammation induces fatigue behavior are not known. This lack of knowledge hinders development of interventions to reduce or prevent this disabling symptom. Infection induced fatigue/lethargy in rodents is mediated by suppression of hypothalamic orexin activity. Orexin is critical for maintaining wakefulness and motivated behavior. Though there are differences between infection and cytotoxic chemotherapy in some symptoms, both induce peripheral inflammation and fatigue. Based on these similarities we hypothesized that cytotoxic chemotherapy induces fatigue by disrupting orexin neuron activity. We found that a single dose of a cytotoxic chemotherapy cocktail (cyclophosphamide, adriamycin, 5-fluorouracil - CAF) induced fatigue/lethargy in mice and rats as evidenced by a significant decline in voluntary locomotor activity measured by telemetry. CAF induced inflammatory gene expression - IL-1R1 (p<0.001), IL-6 (p<0.01), TNFα (p<0.01), and MCP-1 (p<0.05) - in the rodent hypothalamus 6-24h after treatment during maximum fatigue/lethargy. CAF decreased orexin neuron activity as reflected by decreased nuclear cFos localization in orexin neurons 24h after treatment (p<0.05) and by decreased orexin-A in cerebrospinal fluid 16 h after treatment (p<0.001). Most importantly, we found that central administration of 1 μg orexin-A restored activity in CAF-treated rats (p<0.05). These results demonstrate that cytotoxic chemotherapy induces hypothalamic inflammation and that suppression of hypothalamic orexin neuron activity has a causal role in cytotoxic chemotherapy-induced fatigue in rodents.
Collapse
Affiliation(s)
- K B Weymann
- Oregon Health & Science University, Portland, OR 97239, United States.
| | - L J Wood
- Oregon Health & Science University, Portland, OR 97239, United States; School of Nursing, MGH Institute of Health Professions, Boston, MA 02129, United States.
| | - X Zhu
- Oregon Health & Science University, Portland, OR 97239, United States.
| | - D L Marks
- Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
50
|
Urbanski HF, Kohama SG, West GA, Glynn C, Williams-Karnesky RL, Earl E, Neuringer MN, Renner L, Weiss A, Stenzel-Poore M, Bahjat FR. Changes in spontaneous activity assessed by accelerometry correlate with extent of cerebral ischemia-reperfusion injury in the nonhuman primate. Transl Stroke Res 2014; 3:442-51. [PMID: 23580904 DOI: 10.1007/s12975-012-0191-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The use of accelerometry to monitor activity in human stroke patients has revealed strong correlations between objective activity measurements and subjective neurological findings. The goal of our study was to assess the applicability of accelerometry-based measurements in experimental animals undergoing surgically-induced cerebral ischemia. Using a nonhuman primate cortical stroke model, we demonstrate for the first time that monitoring locomotor activity prior to and following cerebrovascular ischemic injury using an accelerometer is feasible in adult male rhesus macaques and that the measured activity outcomes significantly correlate with severity of brain injury. The use of accelerometry as an unobtrusive, objective preclinical efficacy determinant could complement standard practices involving subjective neurological scoring and magnetic resonance imaging in nonhuman primates. Similar activity monitoring devices to those employed in this study are currently in use in human clinical studies, underscoring the feasibility of this approach for assessing the clinical potential of novel treatments for cerebral ischemia.
Collapse
Affiliation(s)
- Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA ; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|