1
|
Hirschberg PR, Siegel DM, Teegala S, Rahbe M, Sarkar P, Routh VH. Reducing neuronal nitric oxide synthase (nNOS) expression in the ventromedial hypothalamus (VMH) increases body weight and blood glucose levels while decreasing physical activity in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594324. [PMID: 38798316 PMCID: PMC11118327 DOI: 10.1101/2024.05.15.594324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Glucose-inhibited (GI) neurons of the ventromedial hypothalamus (VMH) depend on neuronal nitric oxide synthase (nNOS) and AMP-activated protein kinase (AMPK) for activation in low glucose. The Lopez laboratory has shown that the effects of estrogen on brown fat thermogenesis and white fat browning require inhibition of VMH AMPK. This effect of estrogen was mediated by downstream lateral hypothalamus (LH) orexin neurons (1,2). We previously showed that estrogen inhibits activation of GI neurons in low glucose by inhibiting AMPK (3). Thus, we hypothesized that VMH AMPK- and nNOS-dependent GI neurons project to and inhibit orexin neurons. Estrogen inhibition of AMPK in GI neurons would then disinhibit orexin neurons and stimulate brown fat thermogenesis and white fat browning, leading to decreased body weight. To test this hypothesis, we reduced VMH nNOS expression using nNOS shRNA in female mice and measured body weight, adiposity, body temperature, white and brown fat uncoupling protein (UCP1; an index of thermogenesis and browning), locomotor activity, and blood glucose levels. Surprisingly, we saw no effect of reduced VMH nNOS expression on body temperature or UCP1. Instead, body weight and adiposity increased by 30% over 2 weeks post injection of nNOS shRNA. This was associated with increased blood glucose levels and decreased locomotor activity. We also found that VMH nNOS-GI neurons project to the LH. However, stimulation of VMH-LH projections increased excitatory glutamate input onto orexin neurons. Thus, our data do not support our original hypothesis. Excitation of orexin neurons has previously been shown to increase physical activity, leading to decreased blood glucose and body weight (4). We now hypothesize that VMH nNOS-GI neurons play a role in this latter function of orexin neurons.
Collapse
|
2
|
Flores-Ramirez FJ, Illenberger JM, Martin-Fardon R. Interaction between corticotropin-releasing factor, orexin, and dynorphin in the infralimbic cortex may mediate exacerbated alcohol-seeking behavior. Neurobiol Stress 2024; 33:100695. [PMID: 39640001 PMCID: PMC11617300 DOI: 10.1016/j.ynstr.2024.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
A major challenge for the treatment of alcohol use disorder (AUD) is relapse to alcohol use, even after protracted periods of self-imposed abstinence. Stress significantly contributes to the chronic relapsing nature of AUD, given its long-lasting ability to elicit intense craving and precipitate relapse. As individuals transition to alcohol dependence, compensatory allostatic mechanisms result in insults to hypothalamic-pituitary-adrenal axis function, mediated by corticotropin-releasing factor (CRF), which is subsequently hypothesized to alter brain reward pathways, influence affect, elicit craving, and ultimately perpetuate problematic drinking and relapse vulnerability. Orexin (OX; also called hypocretin) plays a well-established role in regulating diverse physiological processes, including stress, and has been shown to interact with CRF. Interestingly, most hypothalamic cells that express Ox mRNA also express Pdyn mRNA. Both dynorphin and OX are located in the same synaptic vesicles, and they are co-released. The infralimbic cortex (IL) of the medial prefrontal cortex (mPFC) has emerged as being directly involved in the compulsive nature of alcohol consumption during dependence. The IL is a CRF-rich region that receives OX projections from the hypothalamus and where OX receptor mRNA has been detected. Although not thoroughly understood, anatomical and behavioral pharmacology data suggest that CRF, OX, and dynorphin may interact, particularly in the IL, and that functional interactions between these three systems in the IL may be critical for the etiology and pervasiveness of compulsive alcohol seeking in dependent subjects that may render them vulnerable to relapse. The present review presents evidence of the role of the IL in AUD and discusses functional interactions between CRF, OX, and dynorphin in this structure and how they are related to exacerbated alcohol drinking and seeking.
Collapse
Affiliation(s)
- Francisco J. Flores-Ramirez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Psychology, California State University, San Marcos, CA, USA
| | | | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
3
|
Bjorness TE, Greene RW. Orexin-mediated motivated arousal and reward seeking. Peptides 2024; 180:171280. [PMID: 39159833 DOI: 10.1016/j.peptides.2024.171280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
4
|
Li Z, Zheng L, Wang J, Wang L, Qi Y, Amin B, Zhu J, Zhang N. Dopamine in the regulation of glucose and lipid metabolism: a narrative review. Obesity (Silver Spring) 2024; 32:1632-1645. [PMID: 39081007 DOI: 10.1002/oby.24068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Owing to the global obesity epidemic, understanding the regulatory mechanisms of glucose and lipid metabolism has become increasingly important. The dopaminergic system, including dopamine, dopamine receptors, dopamine transporters, and other components, is involved in numerous physiological and pathological processes. However, the mechanism of action of the dopaminergic system in glucose and lipid metabolism is poorly understood. In this review, we examine the role of the dopaminergic system in glucose and lipid metabolism. RESULTS The dopaminergic system regulates glucose and lipid metabolism through several mechanisms. It regulates various activities at the central level, including appetite control and decision-making, which contribute to regulating body weight and energy metabolism. In the pituitary gland, dopamine inhibits prolactin production and promotes insulin secretion through dopamine receptor 2. Furthermore, it can influence various physiological components in the peripheral system, such as pancreatic β cells, glucagon-like peptide-1, adipocytes, hepatocytes, and muscle, by regulating insulin and glucagon secretion, glucose uptake and use, and fatty acid metabolism. CONCLUSIONS The role of dopamine in regulating glucose and lipid metabolism has significant implications for the physiology and pathogenesis of disease. The potential therapeutic value of dopamine lies in its effects on metabolic disorders.
Collapse
Affiliation(s)
- Zhehong Li
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lifei Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing Wang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yao Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Buhe Amin
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jinxia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Nengwei Zhang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Mavanji V, Pomonis BL, Shekels L, Kotz CM. Interactions between Lateral Hypothalamic Orexin and Dorsal Raphe Circuitry in Energy Balance. Brain Sci 2024; 14:464. [PMID: 38790443 PMCID: PMC11117928 DOI: 10.3390/brainsci14050464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Orexin/hypocretin terminals innervate the dorsal raphe nucleus (DRN), which projects to motor control areas important for spontaneous physical activity (SPA) and energy expenditure (EE). Orexin receptors are expressed in the DRN, and obesity-resistant (OR) rats show higher expression of these receptors in the DRN and elevated SPA/EE. We hypothesized that orexin-A in the DRN enhances SPA/EE and that DRN-GABA modulates the effect of orexin-A on SPA/EE. We manipulated orexin tone in the DRN either through direct injection of orexin-A or through the chemogenetic activation of lateral-hypothalamic (LH) orexin neurons. In the orexin neuron activation experiment, fifteen minutes prior to the chemogenetic activation of orexin neurons, the mice received either the GABA-agonist muscimol or antagonist bicuculline injected into the DRN, and SPA/EE was monitored for 24 h. In a separate experiment, orexin-A was injected into the DRN to study the direct effect of DRN orexin on SPA/EE. We found that the activation of orexin neurons elevates SPA/EE, and manipulation of GABA in the DRN does not alter the SPA response to orexin neuron activation. Similarly, intra-DRN orexin-A enhanced SPA and EE in the mice. These results suggest that orexin-A in the DRN facilitates negative energy balance by increasing physical activity-induced EE, and that modulation of DRN orexin-A is a potential strategy to promote SPA and EE.
Collapse
Affiliation(s)
- Vijayakumar Mavanji
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (V.M.); (B.L.P.); (L.S.)
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brianna L. Pomonis
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (V.M.); (B.L.P.); (L.S.)
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laurie Shekels
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (V.M.); (B.L.P.); (L.S.)
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Catherine M. Kotz
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (V.M.); (B.L.P.); (L.S.)
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA
- Geriatric Research, Education and Clinical Center, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
| |
Collapse
|
6
|
Xu L, Xue R, Ai Z, Huang Y, Liu X, Wang L, Liang D, Wang Z. Resting-State Functional Magnetic Resonance Imaging as an Indicator of Neuropsychological Changes in Type 1 Narcolepsy. Acad Radiol 2024; 31:69-81. [PMID: 37821344 DOI: 10.1016/j.acra.2023.08.026] [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: 06/29/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 10/13/2023]
Abstract
RATIONALE AND OBJECTIVES To explore indicators of neuropsychological changes in patients with type 1 narcolepsy (NT1) using resting-state functional magnetic resonance imaging (rs-fMRI). MATERIALS AND METHODS Thirty-four NT1 patients and 34 age- and sex-matched healthy volunteers were recruited for neuropsychiatric assessments and rs-fMRI data acquisition. Fractional amplitude of low-frequency fluctuations (fALFF), regional homogeneity (ReHo), and related brain functional connectivity (FC) were calculated for the two groups and compared using a two-sample t test with cluster-level FDR correction. Moreover, partial correlation analysis was performed between these functional values of changed brain regions and clinical scales. RESULTS Compared to those of healthy controls, spontaneous functional activities were significantly weakened in patients with NT1 in regions such as the left/right posterior cerebellum lobe, left inferior temporal gyrus, and left dorsolateral superior frontal gyrus, whereas those in regions such as the left middle occipital gyrus, right inferior occipital gyrus, and left/right lingual gyrus were significantly strengthened. Furthermore, NT1 patients displayed significantly changed FCs between the left/right anterior cingulate gyrus (ACG) and regions such as the left/right cerebellum, left middle occipital gyrus, and left inferior frontal gyrus in the operculum. In partial correlation analysis, the functions in the left dorsolateral superior frontal gyrus were significantly related to the Trail Making Tests (TMT) score. Moreover, the FC between the left ACG and left inferior frontal gyrus in the operculum was highly correlated with anxiety and depression features, including the Hamilton Anxiety Scale (HAMA) score and Hamilton Depression Rating Scale (HAMD-17) score. CONCLUSION Patients with NT1 exhibited abnormalities in the anterior cingulate cortex, frontal-parietal cortex, hippocampus, and left/right posterior cerebellum lobe. The deactivation of the left frontal-temporal cortex is stronger, which is involved in the cognitive decline and mental disorders in these patients. Damage to the ACG may affect its FC with other regions and cause cognition and emotion dysregulation, perhaps by impairing patients' visual pathways and frontal-temporal-parietal networks. Hence, these could be important biomarkers for their neuropsychological changes.
Collapse
Affiliation(s)
- Lin Xu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China (L.X., R.X., Y.H., D.L.)
| | - Rong Xue
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China (L.X., R.X., Y.H., D.L.)
| | - Zhu Ai
- Department of Neurology, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (Z.A.)
| | - Yaqin Huang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China (L.X., R.X., Y.H., D.L.)
| | - Xuan Liu
- Department of Neurology, Airport Hospital, General Hospital of Tianjin Medical University, Tianjin, China (X.L.)
| | - Linlin Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China (L.W.)
| | - Danqi Liang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China (L.X., R.X., Y.H., D.L.)
| | - Zuojun Wang
- Department of Diagnostic Radiology, University of Hong Kong, Hong Kong, China (Z.W.).
| |
Collapse
|
7
|
Busebee B, Ghusn W, Cifuentes L, Acosta A. Obesity: A Review of Pathophysiology and Classification. Mayo Clin Proc 2023; 98:1842-1857. [PMID: 37831039 PMCID: PMC10843116 DOI: 10.1016/j.mayocp.2023.05.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 10/14/2023]
Abstract
Obesity is a chronic, multifactorial, and morbid disease. In the United States, 69% of adults are overweight or have obesity, and the global prevalence of obesity is increasing. Obesity is influenced by genetic, neurologic, metabolic, enteric, and behavioral processes. It remains a key modifiable risk factor for many comorbid diseases, including cardiovascular disease, diabetes mellitus, and cancer. Whereas there are recent and significant advances in obesity therapy, including diets, lifestyle modifications, pharmacotherapies, endoscopic procedures, and bariatric surgeries, there is an immense need for a better understanding of the heterogeneity in the pathophysiologic process of obesity and outcomes. Here we review salient pathophysiologic mechanisms underlying the development and morbidity of obesity as well as pathophysiologically based classification systems that inform current obesity management and may inform improved and individualized management in the future.
Collapse
Affiliation(s)
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
8
|
Illenberger JM, Flores-Ramirez FJ, Pascasio G, Matzeu A, Martin-Fardon R. Daily treatment with the dual orexin receptor antagonist DORA-12 during oxycodone abstinence decreases oxycodone conditioned reinstatement. Neuropharmacology 2023; 239:109685. [PMID: 37579870 PMCID: PMC10529002 DOI: 10.1016/j.neuropharm.2023.109685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
Chronic opioid use disturbs circadian rhythm and sleep, encouraging opioid use and relapse. The orexin (OX) system is recruited by opioids and regulates physiological processes including sleep. Dual OX receptor antagonists (DORAs), developed for insomnia treatment, may relieve withdrawal-associated sleep disturbances. This study investigated whether DORA-12, a recently developed DORA, reduces physiological activity disturbances during oxycodone abstinence and consequently prevents oxycodone-seeking behavior. Male and female Wistar rats were trained to intravenously self-administer oxycodone (0.15 mg/kg, 21 sessions; 8 h/session) in the presence of a contextual/discriminative stimulus (SD). The rats were subsequently housed individually (22 h/day) to monitor activity, food and water intake. They received DORA-12 (0-30 mg/kg, p.o.) after undergoing daily 1-h extinction training (14 days). After extinction, the rats were tested for oxycodone-seeking behavior elicited by the SD. Hypothalamus sections were processed to assess oxycodone- or DORA-12-associated changes to the OX cell number. In males, oxycodone-associated increases in activity during the light-phase, reinstatement, and decreases in the number of OX cells observed in the vehicle-treated group were not observed with DORA-12-treatment. Oxycodone-associated increases in light-phase food and water intake were not observed by day 14 of 3 mg/kg DORA-12-treatment and dark-phase water intake was increased across treatment days. In females, OX cell number was unaffected by oxycodone or DORA-12. Three and 30 mg/kg DORA-12 increased females' day 7 dark-phase activity and decreased reinstatement. Thirty mg/kg DORA-12 reduced oxycodone-associated increases in light-phase food and water intake. The results suggest that DORA-12 improves oxycodone-induced disruptions to physiological activities and reduces relapse.
Collapse
Affiliation(s)
- Jessica M Illenberger
- The Scripps Research Institute, 10550 North Torrey Pines Road, SR-107, La Jolla, CA, 92037, USA.
| | | | - Glenn Pascasio
- The Scripps Research Institute, 10550 North Torrey Pines Road, SR-107, La Jolla, CA, 92037, USA
| | - Alessandra Matzeu
- The Scripps Research Institute, 10550 North Torrey Pines Road, SR-107, La Jolla, CA, 92037, USA
| | - Rémi Martin-Fardon
- The Scripps Research Institute, 10550 North Torrey Pines Road, SR-107, La Jolla, CA, 92037, USA
| |
Collapse
|
9
|
Sa M, Yoo ES, Koh W, Park MG, Jang HJ, Yang YR, Bhalla M, Lee JH, Lim J, Won W, Kwon J, Kwon JH, Seong Y, Kim B, An H, Lee SE, Park KD, Suh PG, Sohn JW, Lee CJ. Hypothalamic GABRA5-positive neurons control obesity via astrocytic GABA. Nat Metab 2023; 5:1506-1525. [PMID: 37653043 DOI: 10.1038/s42255-023-00877-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
The lateral hypothalamic area (LHA) regulates food intake and energy balance. Although LHA neurons innervate adipose tissues, the identity of neurons that regulate fat is undefined. Here we show that GABRA5-positive neurons in LHA (GABRA5LHA) polysynaptically project to brown and white adipose tissues in the periphery. GABRA5LHA are a distinct subpopulation of GABAergic neurons and show decreased pacemaker firing in diet-induced obesity mouse models in males. Chemogenetic inhibition of GABRA5LHA suppresses fat thermogenesis and increases weight gain, whereas gene silencing of GABRA5 in LHA decreases weight gain. In the diet-induced obesity mouse model, GABRA5LHA are tonically inhibited by nearby reactive astrocytes releasing GABA, which is synthesized by monoamine oxidase B (Maob). Gene silencing of astrocytic Maob in LHA facilitates fat thermogenesis and reduces weight gain significantly without affecting food intake, which is recapitulated by administration of a Maob inhibitor, KDS2010. We propose that firing of GABRA5LHA suppresses fat accumulation and selective inhibition of astrocytic GABA is a molecular target for treating obesity.
Collapse
Affiliation(s)
- Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Mingu Gordon Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hyun-Jun Jang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yong Ryoul Yang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Joon-Ho Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yejin Seong
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Heeyoung An
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Pann-Ghill Suh
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea.
- IBS School, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
10
|
Bouâouda H, Jha PK. Orexin and MCH neurons: regulators of sleep and metabolism. Front Neurosci 2023; 17:1230428. [PMID: 37674517 PMCID: PMC10478345 DOI: 10.3389/fnins.2023.1230428] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Sleep-wake and fasting-feeding are tightly coupled behavioral states that require coordination between several brain regions. The mammalian lateral hypothalamus (LH) is a functionally and anatomically complex brain region harboring heterogeneous cell populations that regulate sleep, feeding, and energy metabolism. Significant attempts were made to understand the cellular and circuit bases of LH actions. Rapid advancements in genetic and electrophysiological manipulation help to understand the role of discrete LH cell populations. The opposing action of LH orexin/hypocretin and melanin-concentrating hormone (MCH) neurons on metabolic sensing and sleep-wake regulation make them the candidate to explore in detail. This review surveys the molecular, genetic, and neuronal components of orexin and MCH signaling in the regulation of sleep and metabolism.
Collapse
Affiliation(s)
- Hanan Bouâouda
- Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Pawan Kumar Jha
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
11
|
Gangitano E, Martinez-Sanchez N, Bellini MI, Urciuoli I, Monterisi S, Mariani S, Ray D, Gnessi L. Weight Loss and Sleep, Current Evidence in Animal Models and Humans. Nutrients 2023; 15:3431. [PMID: 37571368 PMCID: PMC10420950 DOI: 10.3390/nu15153431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Sleep is a vital process essential for survival. The trend of reduction in the time dedicated to sleep has increased in industrialized countries, together with the dramatic increase in the prevalence of obesity and diabetes. Short sleep may increase the risk of obesity, diabetes and cardiovascular disease, and on the other hand, obesity is associated with sleep disorders, such as obstructive apnea disease, insomnia and excessive daytime sleepiness. Sleep and metabolic disorders are linked; therefore, identifying the physiological and molecular pathways involved in sleep regulation and metabolic homeostasis can play a major role in ameliorating the metabolic health of the individual. Approaches aimed at reducing body weight could provide benefits for both cardiometabolic risk and sleep quality, which indirectly, in turn, may determine an amelioration of the cardiometabolic phenotype of individuals. We revised the literature on weight loss and sleep, focusing on the mechanisms and the molecules that may subtend this relationship in humans as in animal models.
Collapse
Affiliation(s)
- Elena Gangitano
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Noelia Martinez-Sanchez
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | | | - Irene Urciuoli
- Department of Surgery, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Monterisi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Stefania Mariani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - David Ray
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
12
|
Song XJ, Yang CL, Chen D, Yang Y, Mao Y, Cao P, Jiang A, Wang W, Zhang Z, Tao W. Up-regulation of LCN2 in the anterior cingulate cortex contributes to neural injury-induced chronic pain. Front Cell Neurosci 2023; 17:1140769. [PMID: 37362002 PMCID: PMC10285483 DOI: 10.3389/fncel.2023.1140769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
Chronic pain caused by disease or injury affects more than 30% of the general population. The molecular and cellular mechanisms underpinning the development of chronic pain remain unclear, resulting in scant effective treatments. Here, we combined electrophysiological recording, in vivo two-photon (2P) calcium imaging, fiber photometry, Western blotting, and chemogenetic methods to define a role for the secreted pro-inflammatory factor, Lipocalin-2 (LCN2), in chronic pain development in mice with spared nerve injury (SNI). We found that LCN2 expression was upregulated in the anterior cingulate cortex (ACC) at 14 days after SNI, resulting in hyperactivity of ACC glutamatergic neurons (ACCGlu) and pain sensitization. By contrast, suppressing LCN2 protein levels in the ACC with viral constructs or exogenous application of neutralizing antibodies leads to significant attenuation of chronic pain by preventing ACCGlu neuronal hyperactivity in SNI 2W mice. In addition, administering purified recombinant LCN2 protein in the ACC could induce pain sensitization by inducing ACCGlu neuronal hyperactivity in naïve mice. This study provides a mechanism by which LCN2-mediated hyperactivity of ACCGlu neurons contributes to pain sensitization, and reveals a new potential target for treating chronic pain.
Collapse
Affiliation(s)
- Xiang-Jie Song
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen-Ling Yang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Danyang Chen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yumeng Yang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu Mao
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Peng Cao
- Department of Neurology, Stroke Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Aijun Jiang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenjuan Tao
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Purnell JQ. What is Obesity?: Definition as a Disease, with Implications for Care. Gastroenterol Clin North Am 2023; 52:261-275. [PMID: 37197872 DOI: 10.1016/j.gtc.2023.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Advances in the understanding of weight regulation provide the framework for the recognition of obesity as a chronic disease. Lifestyle approaches are foundational in the prevention of obesity and should be continued while weight management interventions, including antiobesity medications and metabolic-bariatric procedures, are offered to eligible patients. Clinical challenges remain, however, including overcoming obesity stigma and bias within the medical community toward medical and surgical approaches, ensuring insurance coverage for obesity management (including medications and surgery), and promoting policies that reverse the upward worldwide trend in obesity and adiposity complications in populations.
Collapse
Affiliation(s)
- Jonathan Q Purnell
- Knight Cardiovascular Institute and Division of Endocrinology, Diabetes, and Clinical Nutrition, Oregon Health & Science University, Mailcode: HRC5N, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
14
|
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
|
15
|
Illenberger JM, Flores-Ramirez FJ, Matzeu A, Mason BJ, Martin-Fardon R. Suvorexant, an FDA-approved dual orexin receptor antagonist, reduces oxycodone self-administration and conditioned reinstatement in male and female rats. Front Pharmacol 2023; 14:1127735. [PMID: 37180716 PMCID: PMC10172671 DOI: 10.3389/fphar.2023.1127735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/17/2023] [Indexed: 05/16/2023] Open
Abstract
Background: The Department of Health and Human Services reports that prescription pain reliever (e.g., oxycodone) misuse was initiated by 4,400 Americans each day in 2019. Amid the opioid crisis, effective strategies to prevent and treat prescription opioid use disorder (OUD) are pressing. In preclinical models, the orexin system is recruited by drugs of abuse, and blockade of orexin receptors (OX receptors) prevents drug-seeking behavior. The present study sought to determine whether repurposing suvorexant (SUV), a dual OX receptor antagonist marketed for the treatment of insomnia, can treat two features of prescription OUD: exaggerated consumption and relapse. Methods: Male and female Wistar rats were trained to self-administer oxycodone (0.15 mg/kg, i. v., 8 h/day) in the presence of a contextual/discriminative stimulus (SD) and the ability of SUV (0-20 mg/kg, p. o.) to decrease oxycodone self-administration was tested. After self-administration testing, the rats underwent extinction training, after which we tested the ability of SUV (0 and 20 mg/kg, p. o.) to prevent reinstatement of oxycodone seeking elicited by the SD. Results: The rats acquired oxycodone self-administration and intake was correlated with the signs of physical opioid withdrawal. Additionally, females self-administered approximately twice as much oxycodone as males. Although SUV had no overall effect on oxycodone self-administration, scrutiny of the 8-h time-course revealed that 20 mg/kg SUV decreased oxycodone self-administration during the first hour in males and females. The oxycodone SD elicited strong reinstatement of oxycodone-seeking behavior that was significantly more robust in females. Suvorexant blocked oxycodone seeking in males and reduced it in females. Conclusions: These results support the targeting of OX receptors for the treatment for prescription OUD and repurposing SUV as pharmacotherapy for OUD.
Collapse
|
16
|
Dai Z, Liu Y, Nie L, Chen W, Xu X, Li Y, Zhang J, Shen F, Sui N, Liang J. Locus coeruleus input-modulated reactivation of dentate gyrus opioid-withdrawal engrams promotes extinction. Neuropsychopharmacology 2023; 48:327-340. [PMID: 36302846 PMCID: PMC9751301 DOI: 10.1038/s41386-022-01477-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 12/26/2022]
Abstract
Extinction training during the reconsolidation window following memory recall is an effective behavioral pattern for promoting the extinction of pathological memory. However, promoted extinction by recall-extinction procedure has not been universally replicated in different studies. One potential reason for this may relate to whether initially acquired memory is successfully activated. Thus, the methods for inducing the memory into an active or plastic condition may contribute to promoting its extinction. The aim of this study is to find and demonstrate a manipulatable neural circuit that engages in the memory recall process and where its activation improves the extinction process through recall-extinction procedure. Here, naloxone-precipitated conditioned place aversion (CPA) in morphine-dependent mice was mainly used as a pathological memory model. We found that the locus coeruleus (LC)-dentate gyrus (DG) circuit was necessary for CPA memory recall and that artificial activation of LC inputs to the DG just prior to initiating a recall-extinction procedure significantly promoted extinction learning. We also found that activating this circuit caused an increase in the ensemble size of DG engram cells activated during the extinction, which was confirmed by a cFos targeted strategy to label cells combined with immunohistochemical and in vivo calcium imaging techniques. Collectively, our data uncover that the recall experience is important for updating the memory during the reconsolidation window; they also suggest a promising neural circuit or target based on the recall-extinction procedure for weakening pathological aversion memory, such as opioid withdrawal memory and fear memory.
Collapse
Affiliation(s)
- Zhonghua Dai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Liu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Lina Nie
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Weiqi Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Xing Xu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fang Shen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
17
|
Dhafar HO, BaHammam AS. Body Weight and Metabolic Rate Changes in Narcolepsy: Current Knowledge and Future Directions. Metabolites 2022; 12:1120. [PMID: 36422261 PMCID: PMC9693066 DOI: 10.3390/metabo12111120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 08/26/2023] Open
Abstract
Narcolepsy is a known auto-immune disease that presents mainly in the teenage years with irresistible sleep attacks. Patients with narcolepsy, especially NT1, have been found to have a high prevalence of obesity and other metabolic derangements. This narrative review aimed to address the relationship between narcolepsy and changes in weight and metabolic rate, and discuss potential mechanisms for weight gain and metabolic changes and future research agendas on this topic. This article will provide a balanced, up-to-date critical review of the current literature, and delineate areas for future research, in order to understand the pathophysiological metabolic changes in narcolepsy. Articles using predefined keywords were searched for in PubMed and Google Scholar databases, with predefined inclusion and exclusion criteria. Compared to controls, patients with narcolepsy are more likely to be obese and have higher BMIs and waist circumferences. According to recent research, weight gain in narcolepsy patients may be higher during the disease's outset. The precise mechanisms causing this weight gain remains unknown. The available information, albeit limited, does not support differences in basal or resting metabolic rates between patients with narcolepsy and controls, other than during the time of disease onset. The evidence supporting the role of orexin in weight gain in humans with narcolepsy is still controversial, in the literature. Furthermore, the available data did not show any appreciable alterations in the levels of CSF melanin-concentrating hormone, plasma and CSF leptin, or serum growth hormone, in relation to weight gain. Other mechanisms have been proposed, including a reduction in sympathetic tone, hormonal changes, changes in eating behavior and physical activity, and genetic predisposition. The association between increased body mass index and narcolepsy is well-recognized; however, the relationship between narcolepsy and other metabolic measures, such as body fat/muscle distribution and metabolic rate independent of BMI, is not well documented, and the available evidence is inconsistent. Future longitudinal studies with larger sample sizes are needed to assess BMR in patients with narcolepsy under a standard protocol at the outset of narcolepsy, with regular follow-up.
Collapse
Affiliation(s)
- Hamza O. Dhafar
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Family Medicine, Prince Mansour Military Hospital, Taif 26526, Saudi Arabia
| | - Ahmed S. BaHammam
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
- The Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, P.O. Box 2454, Riyadh 11324, Saudi Arabia
| |
Collapse
|
18
|
Bingul A, Merlin S, Carrive P, Killcross S, Furlong TM. Targeting the lateral hypothalamus with short hairpin RNAs reduces habitual behaviour following extended instrumental training in rats. Neurobiol Learn Mem 2022; 193:107657. [DOI: 10.1016/j.nlm.2022.107657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 10/17/2022]
|
19
|
Le N, Sayers S, Mata-Pacheco V, Wagner EJ. The PACAP Paradox: Dynamic and Surprisingly Pleiotropic Actions in the Central Regulation of Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:877647. [PMID: 35721722 PMCID: PMC9198406 DOI: 10.3389/fendo.2022.877647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP), a pleiotropic neuropeptide, is widely distributed throughout the body. The abundance of PACAP expression in the central and peripheral nervous systems, and years of accompanying experimental evidence, indicates that PACAP plays crucial roles in diverse biological processes ranging from autonomic regulation to neuroprotection. In addition, PACAP is also abundantly expressed in the hypothalamic areas like the ventromedial and arcuate nuclei (VMN and ARC, respectively), as well as other brain regions such as the nucleus accumbens (NAc), bed nucleus of stria terminalis (BNST), and ventral tegmental area (VTA) - suggesting that PACAP is capable of regulating energy homeostasis via both the homeostatic and hedonic energy balance circuitries. The evidence gathered over the years has increased our appreciation for its function in controlling energy balance. Therefore, this review aims to further probe how the pleiotropic actions of PACAP in regulating energy homeostasis is influenced by sex and dynamic changes in energy status. We start with a general overview of energy homeostasis, and then introduce the integral components of the homeostatic and hedonic energy balance circuitries. Next, we discuss sex differences inherent to the regulation of energy homeostasis via these two circuitries, as well as the activational effects of sex steroid hormones that bring about these intrinsic disparities between males and females. Finally, we explore the multifaceted role of PACAP in regulating homeostatic and hedonic feeding through its actions in regions like the NAc, BNST, and in particular the ARC, VMN and VTA that occur in sex- and energy status-dependent ways.
Collapse
Affiliation(s)
- Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
20
|
Levine AS, Jewett DC, Kotz CM, Olszewski PK. Behavioral plasticity: Role of neuropeptides in shaping feeding responses. Appetite 2022; 174:106031. [PMID: 35395362 DOI: 10.1016/j.appet.2022.106031] [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: 12/19/2021] [Revised: 03/12/2022] [Accepted: 03/29/2022] [Indexed: 11/28/2022]
Abstract
Behavioral plasticity refers to changes occurring due to external influences on an organism, including adaptation, learning, memory and enduring influences from early life experience. There are 2 types of behavioral plasticity: "developmental", which refers to gene/environment interactions affecting a phenotype, and "activational" which refers to innate physiology and can involve structural physiological changes of the body. In this review, we focus on feeding behavior, and studies involving neuropeptides that influence behavioral plasticity - primarily opioids, orexin, neuropeptide Y, and oxytocin. In each section of the review, we include examples of behavioral plasticity as it relates to actions of these neuropeptides. It can be concluded from this review that eating behavior is influenced by a number of external factors, including time of day, type of food available, energy balance state, and stressors. The reviewed work underscores that environmental factors play a critical role in feeding behavior and energy balance, but changes in eating behavior also result from a multitude of non-environmental factors, such that there can be no single mechanism or variable that can explain ingestive behavior.
Collapse
Affiliation(s)
- Allen S Levine
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55113, USA.
| | - David C Jewett
- Department of Psychology, University of Wisconsin-Eau Claire, Eau Claire, WI, USA
| | - Catherine M Kotz
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, 55414, USA; Geriatric, Research, Education and Clinical Center, Minneapolis Veterans Affairs Health, Minneapolis, MN, 55417, USA
| | - Pawel K Olszewski
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55113, USA; Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, 55414, USA; Faculty of Science and Engineering, University of Waikato, Hamilton, 3240, New Zealand
| |
Collapse
|
21
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
22
|
Tran LT, Park S, Kim SK, Lee JS, Kim KW, Kwon O. Hypothalamic control of energy expenditure and thermogenesis. Exp Mol Med 2022; 54:358-369. [PMID: 35301430 PMCID: PMC9076616 DOI: 10.1038/s12276-022-00741-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Energy expenditure and energy intake need to be balanced to maintain proper energy homeostasis. Energy homeostasis is tightly regulated by the central nervous system, and the hypothalamus is the primary center for the regulation of energy balance. The hypothalamus exerts its effect through both humoral and neuronal mechanisms, and each hypothalamic area has a distinct role in the regulation of energy expenditure. Recent studies have advanced the understanding of the molecular regulation of energy expenditure and thermogenesis in the hypothalamus with targeted manipulation techniques of the mouse genome and neuronal function. In this review, we elucidate recent progress in understanding the mechanism of how the hypothalamus affects basal metabolism, modulates physical activity, and adapts to environmental temperature and food intake changes. The hypothalamus is a key regulator of metabolism, controlling resting metabolism, activity levels, and responses to external temperature and food intake. The balance between energy intake and expenditure must be tightly controlled, with imbalances resulting in metabolic disorders such as obesity or diabetes. Obin Kwon at Seoul National University College of Medicine and Ki Woo Kim at Yonsei University College of Dentistry, Seoul, both in South Korea, and coworkers reviewed how metabolism is regulated by the hypothalamus, a small hormone-producing brain region. They report that hormonal and neuronal signals from the hypothalamus influence the ratio of lean to fatty tissue, gender-based differences in metabolism, activity levels, and weight gain in response to food intake. They note that further studies to untangle cause-and-effect relationships and other genetic factors will improve our understanding of metabolic regulation.
Collapse
Affiliation(s)
- Le Trung Tran
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Sohee Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Departments of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Seul Ki Kim
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Jin Sun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Departments of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Ki Woo Kim
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea.
| | - Obin Kwon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Departments of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
23
|
Doucette WT, Smedley EB, Ruiz-Jaquez M, Khokhar JY, Smith KS. Chronic Chemogenetic Manipulation of Ventral Pallidum Targeted Neurons in Male Rats Fed an Obesogenic Diet. Brain Res 2022; 1784:147886. [DOI: 10.1016/j.brainres.2022.147886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/14/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
|
24
|
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
|
25
|
Vaseghi S, Zarrabian S, Haghparast A. Reviewing the role of the orexinergic system and stressors in modulating mood and reward-related behaviors. Neurosci Biobehav Rev 2021; 133:104516. [PMID: 34973302 DOI: 10.1016/j.neubiorev.2021.104516] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 01/22/2023]
Abstract
In this review study, we aimed to introduce the orexinergic system as an important signaling pathway involved in a variety of cognitive functions such as memory, motivation, and reward-related behaviors. This study focused on the role of orexinergic system in modulating reward-related behavior, with or without the presence of stressors. Cross-talk between the reward system and orexinergic signaling was also investigated, especially orexinergic signaling in the ventral tegmental area (VTA), the nucleus accumbens (NAc), and the hippocampus. Furthermore, we discussed the role of the orexinergic system in modulating mood states and mental illnesses such as depression, anxiety, panic, and posttraumatic stress disorder (PTSD). Here, we narrowed down our focus on the orexinergic signaling in three brain regions: the VTA, NAc, and the hippocampus (CA1 region and dentate gyrus) for their prominent role in reward-related behaviors and memory. It was concluded that the orexinergic system is critically involved in reward-related behavior and significantly alters stress responses and stress-related psychiatric and mood disorders.
Collapse
Affiliation(s)
- Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Shahram Zarrabian
- Department of Anatomical Sciences & Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran.
| |
Collapse
|
26
|
Moslehi E, Minasian V, Sadeghi H. Subcutaneous Adipose Tissue Browning, Serum Orexin-A, and Insulin Resistance Following Aerobic Exercise in High-Fat Diet Obesity Male Wistar Rats. Int J Prev Med 2021; 12:132. [PMID: 34912508 PMCID: PMC8631127 DOI: 10.4103/ijpvm.ijpvm_110_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 12/04/2019] [Indexed: 11/04/2022] Open
Abstract
Background Subcutaneous adipose tissue (SAT) relative to the other adipose tissues may have different roles in health and insulin resistance. The purpose of this study was to investigate the effectiveness of aerobic exercise on SAT thermogenesis indices, serum orexin-A (OXA), and insulin resistance in high-fat diet-induced obesity male Wistar rats. Methods Thirty-two male Wistar rats with an average weight of 180-200 g were randomly assigned into 4 equal groups: normal fat diet (NFD), high-fat diet obesity (HFDO), normal fat diet after high-fat diet obesity (HFDO-NFD), and aerobic exercise group with normal fat diet after high-fat diet obesity (HFDO-AEX). Fasting levels of serum OXA, insulin, FBS, high-density lipoproteins, low-density lipoproteins, cholesterol and gene expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and UCP1 in SAT were evaluated. Samples were taken in the HFDO group after obesity-induced and in other groups 48 h after 8 weeks of aerobic exercise. Results The results showed that HFD significantly decreased serum levels of OXA, HDL-c and gene expression of PGC1α and UCP1 in SAT. In addition, it caused a significant increase in Lee index, FBS, insulin resistance, and serum lipid profile in comparison with the NFD group (P ≤ 0.001). Aerobic exercise significantly modified the changes caused by HFD to the normal levels (P ≤ 0.001). Conclusions These data suggest that aerobic exercise caused an improvement in insulin resistance and blood lipid profiles through an increase in the serum level of OXA and alteration in the SAT phenotype from white to brown or beige.
Collapse
Affiliation(s)
- Ebrahim Moslehi
- Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
| | - Vazgen Minasian
- Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
| | - Heibatollah Sadeghi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, I.R. Iran
| |
Collapse
|
27
|
van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obes Rev 2021; 22:e13210. [PMID: 33559362 PMCID: PMC8243944 DOI: 10.1111/obr.13210] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022]
Abstract
The role of serotonin in food intake has been studied for decades. Food intake is mainly regulated by two brain circuitries: (i) the homeostatic circuitry, which matches energy intake to energy expenditure, and (ii) the hedonic circuitry, which is involved in rewarding and motivational aspects of energy consumption. In the homeostatic circuitry, serotonergic signaling contributes to the integration of metabolic signals that convey the body's energy status and facilitates the ability to suppress food intake when homeostatic needs have been met. In the hedonic circuitry, serotonergic signaling may reduce reward-related, motivational food consumption. In contrast, peripherally acting serotonin promotes energy absorption and storage. Disturbed serotonergic signaling is associated with obesity, emphasizing the importance to understand the role of serotonergic signaling in food intake. However, unraveling the serotonin-mediated regulation of food intake is complex, as the effects of serotonergic signaling in different brain regions depend on the regional expression of serotonin receptor subtypes and downstream effects via connections to other brain regions. We therefore provide an overview of the effects of serotonergic signaling in brain regions of the homeostatic and hedonic regulatory systems on food intake. Furthermore, we discuss the disturbances in serotonergic signaling in obesity and its potential therapeutic implications.
Collapse
Affiliation(s)
- Katy A van Galen
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Muthmainah M, Gogos A, Sumithran P, Brown RM. Orexins (hypocretins): The intersection between homeostatic and hedonic feeding. J Neurochem 2021; 157:1473-1494. [PMID: 33608877 DOI: 10.1111/jnc.15328] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
Orexins are hypothalamic neuropeptides originally discovered to play a role in the regulation of feeding behaviour. The broad connections of orexin neurons to mesocorticolimbic circuitry suggest they may play a role in mediating reward-related behaviour beyond homeostatic feeding. Here, we review the role of orexin in a variety of eating-related behaviour, with a focus on reward and motivation, and the neural circuits driving these effects. One emerging finding is the involvement of orexins in hedonic and appetitive behaviour towards palatable food, in addition to their role in homeostatic feeding. This review discusses the brain circuitry and possible mechanisms underlying the role of orexins in these behaviours. Overall, there is a marked bias in the literature towards studies involving male subjects. As such, future work needs to be done to involve female subjects. In summary, orexins play an important role in driving motivation for high salient rewards such as highly palatable food and may serve as the intersection between homeostatic and hedonic feeding.
Collapse
Affiliation(s)
- Muthmainah Muthmainah
- The Florey Institute of Neuroscience and Mental Health, Mental Health Research Theme, Parkville, Melbourne, Vic., Australia.,The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Vic., Australia.,Department of Anatomy, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Andrea Gogos
- The Florey Institute of Neuroscience and Mental Health, Mental Health Research Theme, Parkville, Melbourne, Vic., Australia
| | - Priya Sumithran
- Department of Medicine (Austin), University of Melbourne, Heidelberg, Vic., Australia.,Department of Endocrinology, Austin Health, Heidelberg, Vic., Australia
| | - Robyn M Brown
- The Florey Institute of Neuroscience and Mental Health, Mental Health Research Theme, Parkville, Melbourne, Vic., Australia.,The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Vic., Australia
| |
Collapse
|
29
|
Effects of exercise and diet intervention on appetite-regulating hormones associated with miRNAs in obese children. Eat Weight Disord 2021; 26:457-465. [PMID: 32072570 DOI: 10.1007/s40519-020-00869-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
AIMS This study investigated the effects of exercise and diet intervention on appetite-regulating hormones and subjective appetite changes in obese children and examined expressions of specific key microRNAs (miRNA, miR). METHODS 16 obese children were included in a training program consisting of exercise and diet intervention for 6 weeks. Before and after the intervention, fasting blood was collected to determine appetite-regulating hormones (leptin, ghrelin, and orexin) and miRNA (miR-103a-3p and miR-200a-3p) levels; eating behavior of the children was reported using the Children Eating Behavior Questionnaire (CEBQ). RESULTS The level of orexin was significantly decreased (P < 0.05), while ghrelin was significantly enhanced (P < 0.05) after 6 weeks. The scores of food responsiveness (FR) and enjoyment of food (EF) of the CEBQ were significantly decreased (P < 0.05) after intervention. The changes of leptin and that of SR were significantly correlated (r = - 0.455, P < 0.05), and the correlation between the alterations of orexin and that of EF was moderate with significance (r = 0.625, P < 0.05). miR-103a-3p expression was not statistically changed, while miR-200a-3p was significantly inhibited after 6-week intervention (P < 0.05). The correlation between relative changes of miR-103a-3p and that of leptin and orexin were both with significant difference (r = 0.413, P < 0.05; r = 0.409, P < 0.05), whereas the alterations of miR-200a-3p were not correlative with hormones or appetite sensation. CONCLUSION Exercise combined with diet intervention for 6 weeks was effective in regulating appetite sensations and hormones in obese children, and miR-103a-3p and miR-200a-3p might provide a foundation for target biomarkers of appetite trait in modulating the energy balance control by exercise and dietary intervention. LEVEL OF EVIDENCE Level III, case-control analytic study. TRIAL REGISTRATION The trial was registered in ClinicalTrials.gov (NCT03762629).
Collapse
|
30
|
Stanojlovic M, Pallais JP, Kotz CM. Inhibition of Orexin/Hypocretin Neurons Ameliorates Elevated Physical Activity and Energy Expenditure in the A53T Mouse Model of Parkinson's Disease. Int J Mol Sci 2021; 22:E795. [PMID: 33466831 PMCID: PMC7830608 DOI: 10.3390/ijms22020795] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
Aside from the classical motor symptoms, Parkinson's disease also has various non-classical symptoms. Interestingly, orexin neurons, involved in the regulation of exploratory locomotion, spontaneous physical activity, and energy expenditure, are affected in Parkinson's. In this study, we hypothesized that Parkinson's-disease-associated pathology affects orexin neurons and therefore impairs functions they regulate. To test this, we used a transgenic animal model of Parkinson's, the A53T mouse. We measured body composition, exploratory locomotion, spontaneous physical activity, and energy expenditure. Further, we assessed alpha-synuclein accumulation, inflammation, and astrogliosis. Finally, we hypothesized that chemogenetic inhibition of orexin neurons would ameliorate observed impairments in the A53T mice. We showed that aging in A53T mice was accompanied by reductions in fat mass and increases in exploratory locomotion, spontaneous physical activity, and energy expenditure. We detected the presence of alpha-synuclein accumulations in orexin neurons, increased astrogliosis, and microglial activation. Moreover, loss of inhibitory pre-synaptic terminals and a reduced number of orexin cells were observed in A53T mice. As hypothesized, this chemogenetic intervention mitigated the behavioral disturbances induced by Parkinson's disease pathology. This study implicates the involvement of orexin in early Parkinson's-disease-associated impairment of hypothalamic-regulated physiological functions and highlights the importance of orexin neurons in Parkinson's disease symptomology.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany
| | - Jean Pierre Pallais
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, 321 Church St SE, Minneapolis, MN 55455, USA; (J.P.P.); (C.M.K.)
| | - Catherine M. Kotz
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, 321 Church St SE, Minneapolis, MN 55455, USA; (J.P.P.); (C.M.K.)
- Minneapolis VA Health Care System, GRECC, 1 Veterans Dr, Minneapolis, MN 55417, USA
| |
Collapse
|
31
|
Synchronous neuronal interactions in rat hypothalamic culture: a novel model for the study of network dynamics in metabolic disorders. Exp Brain Res 2021; 239:755-764. [PMID: 33388905 DOI: 10.1007/s00221-020-05977-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022]
Abstract
Synchronous neural activity is a feature of normal brain function, and altered synchronization is observed in several neurological diseases. Dysfunction in hypothalamic pathways leads to obesity, suggesting that hypothalamic neural synchrony is critical for energy homeostasis. The lateral hypothalamic orexin neurons are extensively interconnected with other brain structures and are important for energy balance. Earlier studies show that rats with higher orexin sensitivity are obesity resistant. Similarly, topiramate, an anti-epileptic drug, has been shown to reduce weight in humans. Since orexin enhances neuronal excitation, we hypothesized that obesity-resistant rats with higher orexin sensitivity may exhibit enhanced hypothalamic synchronization. We further hypothesized that anti-obesity agents such as orexin and topiramate will enhance hypothalamic synchronization. To test this, we examined neural synchronicity in primary embryonic hypothalamic cell cultures, obtained from embryonic day 18 (E18) obesity-susceptible Sprague-Dawley (SD) and obesity-resistant rats. Hypothalamic tissue was cultured in multielectrode array (MEA), and recordings were performed twice weekly, from 4th to 32nd day in vitro (DIV). Next, we tested the effects of orexin and topiramate application on neural synchronicity of hypothalamic cultures obtained from SD rat embryos. Signals were analyzed for synchronization using cross correlation. Our results showed that (1) obesity-resistant hypothalamus exhibits significantly higher synchronization compared to obesity-sensitive hypothalamus; and (2) orexin and topiramate enhance hypothalamic synchronization. These results support that enhanced orexin sensitivity is associated with greater neural synchronization, and that anti-obesity treatments enhance network synchronization, thus constrain variability in hypothalamic output signals, to extrahypothalamic structures involved in energy homeostasis.
Collapse
|
32
|
Obesity and Related Type 2 Diabetes: A Failure of the Autonomic Nervous System Controlling Gastrointestinal Function? GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The pandemic spread of obesity and type 2 diabetes is a serious health problem that cannot be contained with common therapies. At present, the most effective therapeutic tool is metabolic surgery, which substantially modifies the gastrointestinal anatomical structure. This review reflects the state of the art research in obesity and type 2 diabetes, describing the probable reason for their spread, how the various brain sectors are involved (with particular emphasis on the role of the vagal system controlling different digestive functions), and the possible mechanisms for the effectiveness of bariatric surgery. According to the writer’s interpretation, the identification of drugs that can modulate the activity of some receptor subunits of the vagal neurons and energy-controlling structures of the central nervous system (CNS), and/or specific physical treatment of cortical areas, could reproduce, non-surgically, the positive effects of metabolic surgery.
Collapse
|
33
|
Zegarra‐Valdivia JA, Pignatelli J, Fernandez de Sevilla ME, Fernandez AM, Munive V, Martinez‐Rachadell L, Nuñez A, Torres Aleman I. Insulin‐like growth factor I modulates sleep through hypothalamic orexin neurons. FASEB J 2020; 34:15975-15990. [DOI: 10.1096/fj.202001281rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/30/2020] [Accepted: 09/21/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Jonathan A. Zegarra‐Valdivia
- Functional and Systems Neurobiology Department Cajal Institute (CSIC) Madrid Spain
- CIBERNED Madrid Spain
- Universidad Nacional de San Agustín de Arequipa Perú
| | - Jaime Pignatelli
- Functional and Systems Neurobiology Department Cajal Institute (CSIC) Madrid Spain
- CIBERNED Madrid Spain
| | | | - Ana M. Fernandez
- Functional and Systems Neurobiology Department Cajal Institute (CSIC) Madrid Spain
- CIBERNED Madrid Spain
| | - Victor Munive
- Functional and Systems Neurobiology Department Cajal Institute (CSIC) Madrid Spain
- CIBERNED Madrid Spain
| | - Laura Martinez‐Rachadell
- Functional and Systems Neurobiology Department Cajal Institute (CSIC) Madrid Spain
- CIBERNED Madrid Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neuroscience School of Medicine UAM Madrid Spain
| | - Ignacio Torres Aleman
- Functional and Systems Neurobiology Department Cajal Institute (CSIC) Madrid Spain
- CIBERNED Madrid Spain
| |
Collapse
|
34
|
Su M, Yan M, Yao J, Fang Y, Jin H, Gong Y. Unacylated Ghrelin Regulates Glucose-Sensitive Neurons Activity and Glycolipid Metabolism via Orexin-A Neurons in the Lateral Hypothalamic Area. Horm Metab Res 2020; 52:747-754. [PMID: 32731263 DOI: 10.1055/a-1207-1212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The objective of the study was to investigate the regulatory actions of unacylated ghrelin (UAG) on glucose-sensitive (GS) neurons and glycolipid metabolism in the lateral hypothalamus area (LHA) and its involvement with orexin-A-immunopositive neurons. The effects of UAG administered into the LHA on GS neurons discharges and glycolipid metabolism were detected by single neuron discharge recording, biochemical index analysis and quantitative real-time PCR; the level of c-fos protein in orexin-A-immunopositive neurons was observed using immunofluorescence staining. UAG microinjected into the LHA activated glucose-inhibited neurons, which were partially blocked by pre-administration of anti-orexin-A antibody in the LHA. Furthermore, UAG microinjected into the LHA significantly reduced serum triglycerides (TG), total cholesterol, low-density lipoprotein cholesterol, blood glucose, insulin and hepatic TG levels, while elevated serum high-density lipoprotein cholesterol levels. UAG elevated the mRNA expression of carnitine palmitoyltransferase-1 and reduced the mRNA expression of acetyl-CoA carboxylase-1 in the liver. The above-mentioned effects of UAG were partially blocked by pre-administration of anti-orexin-A antibody. The expressions of orexin-A and c-fos were observed in the LHA. After UAG injection into the LHA, some neurons showed double labeling, and the percentage of double-labeled orexin-A/c-fos neurons in orexin-A-immunopositive neurons increased significantly. UAG in the LHA regulates glycolipid metabolism by activating orexin-A-immunopositive neurons in the LHA.
Collapse
Affiliation(s)
- Manqing Su
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Meixing Yan
- Qingdao Women and Children's Hospital, Qingdao, China
| | - Jiatong Yao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yanpeng Fang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Hong Jin
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
35
|
O'Neal TJ, Nooney MN, Thien K, Ferguson SM. Chemogenetic modulation of accumbens direct or indirect pathways bidirectionally alters reinstatement of heroin-seeking in high- but not low-risk rats. Neuropsychopharmacology 2020; 45:1251-1262. [PMID: 31747681 PMCID: PMC7297977 DOI: 10.1038/s41386-019-0571-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 12/29/2022]
Abstract
Opioid addiction has been declared a public health emergency, with fatal overdoses following relapse reaching epidemic proportions and disease-associated costs continuing to escalate. Relapse is often triggered by re-exposure to drug-associated cues, and though the neural substrates responsible for relapse in vulnerable individuals remains ambiguous, the nucleus accumbens (NAc) has been shown to play a central role. NAc direct and indirect pathway medium spiny neurons (dMSNs and iMSNs) can have oppositional control over reward-seeking and associative learning and are critically involved in reinstatement of psychostimulant-seeking. However, whether these pathways similarly regulate reinstatement of opioid-seeking remains unknown, as is their role in modulating motivation to take opioids. Here, we describe a method for classifying addiction severity in outbred rats following intermittent-access heroin self-administration that identifies subgroups as addiction-vulnerable (high-risk) or addiction-resistant (low-risk). Using dual viral-mediated gene transfer of DREADDs, we show that transient inactivation of dMSNs or activation of iMSNs is capable of suppressing cue-induced reinstatement of heroin-seeking in high- but not low-risk rats. Surprisingly, however, the motivation to self-administer heroin was unchanged, indicating a divergence in the encoding of heroin-taking and heroin-seeking in rats. We further show that transient activation of dMSNs or inactivation of iMSNs exacerbates cue-induced reinstatement of heroin-seeking in high- but not low-risk rats, again with no effect on motivation. These findings demonstrate a critical role for dMSNs and iMSNs in encoding vulnerability to reinstatement of heroin-seeking and provide insight into the specific neurobiological changes that occur in vulnerable groups following heroin self-administration.
Collapse
Affiliation(s)
- Timothy J O'Neal
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, 98195, USA
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA, 98195, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Marlaena N Nooney
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Katie Thien
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Susan M Ferguson
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, 98195, USA.
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA, 98195, USA.
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
- Alcohol and Drug Abuse Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
36
|
Linehan V, Fang LZ, Parsons MP, Hirasawa M. High-fat diet induces time-dependent synaptic plasticity of the lateral hypothalamus. Mol Metab 2020; 36:100977. [PMID: 32277924 PMCID: PMC7170999 DOI: 10.1016/j.molmet.2020.100977] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/28/2020] [Accepted: 03/10/2020] [Indexed: 11/15/2022] Open
Abstract
Objective Orexin (ORX) and melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus are critical regulators of energy homeostasis and are thought to differentially contribute to diet-induced obesity. However, it is unclear whether the synaptic properties of these cells are altered by obesogenic diets over time. Methods Rats and mice were fed a control chow or palatable high-fat diet (HFD) for various durations and then synaptic properties of ORX and MCH neurons were examined using exvivo whole-cell patch clamp recording. Confocal imaging was performed to assess the number of excitatory synaptic contacts to these neurons. Results ORX neurons exhibited a transient increase in spontaneous excitatory transmission as early as 1 day up to 1 week of HFD, which returned to control levels with prolonged feeding. Conversely, HFD induced a delayed increase in excitatory synaptic transmission to MCH neurons, which progressively increased as HFD became chronic. This increase occurred before the onset of significant weight gain. These synaptic changes appeared to be due to altered postsynaptic sensitivity or the number of active synaptic contacts depending on cell type and feeding duration. However, HFD induced no change in inhibitory transmission in either cell type at any time point. Conclusions These results suggest that the effects of HFD on feeding-related neurons are cell type-specific and dynamic. This highlights the importance of considering the feeding duration for research and weight loss interventions. ORX neurons may contribute to early hyperphagia, whereas MCH neurons may play a role in the onset and long-term maintenance of diet-induced obesity. High-fat diet increases excitatory transmission to orexin and MCH neurons. Increased excitatory drive to orexin neurons occurs within the first week but is transient. Excitatory synapses to MCH neurons increase with prolonged high-fat diet. Excitatory changes in MCH neurons are delayed but precede significant weight gain. These synaptic changes may contribute to the development and the maintenance of obesity.
Collapse
Affiliation(s)
- Victoria Linehan
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada
| | - Lisa Z Fang
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, A1B 3V6, Canada.
| |
Collapse
|
37
|
Stanojlovic M, Pallais JP, Lee MK, Kotz CM. Pharmacological and chemogenetic orexin/hypocretin intervention ameliorates Hipp-dependent memory impairment in the A53T mice model of Parkinson's disease. Mol Brain 2019; 12:87. [PMID: 31666100 PMCID: PMC6822428 DOI: 10.1186/s13041-019-0514-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/18/2019] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD), classically defined as a progressive motor disorder accompanied with dopaminergic neuron loss and presence of Lewy bodies, is the second most common neurodegenerative disease. PD also has various non-classical symptoms, including cognitive impairments. In addition, inflammation and astrogliosis are recognized as an integral part of PD pathology. The hippocampus (Hipp) is a brain region involved in cognition and memory, and the neuropeptide orexin has been shown to enhance learning and memory. Previous studies show impairments in Hipp-dependent memory in a transgenic mouse model of Parkinson's disease (A53T mice), and we hypothesized that increasing orexin tone will reverse this. To test this, we subjected 3, 5, and 7-month old A53T mice to a Barnes maze and a contextual object recognition test to determine Hipp dependent memory. Inflammation and astrogliosis markers in the Hipp were assessed by immuno-fluorescence densitometry. The data show that early cognitive impairment is coupled with an increase in expression of inflammatory and astrogliosis markers. Next, in two separate experiments, mice were given intra-hippocampal injections of orexin or chemogenetic viral injections of an orexin neuron specific Designer Receptor Exclusively Activated by Designer Drug (DREADD). For the pharmacological approach mice were intracranially treated with orexin A, whereas the chemogenetic approach utilized clozapine N-oxide (CNO). Both pharmacological orexin A intervention as well as chemogenetic activation of orexin neurons ameliorated Hipp-dependent early memory impairment observed in A53T mice. This study implicates orexin in PD-associated cognitive impairment and suggests that exogenous orexin treatment and/or manipulation of endogenous orexin levels may be a potential strategy for addressing early cognitive loss in PD.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA.
| | - Jean Pierre Pallais
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience (ITN), University of Minnesota, Minneapolis, MN, USA
| | - Catherine M Kotz
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
- Minneapolis VA Health Care System, GRECC, Minneapolis, MN, USA
| |
Collapse
|
38
|
Effects of a Flavonoid-Rich Extract from Citrus sinensis Juice on a Diet-Induced Obese Zebrafish. Int J Mol Sci 2019; 20:ijms20205116. [PMID: 31619003 PMCID: PMC6834169 DOI: 10.3390/ijms20205116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Obesity is a pathological condition that has reached epidemic proportions; hence, it is necessary to find novel strategies aimed at fighting this disease. The present study was designed to evaluate the effect of a flavonoid-rich extract of orange (Citrus sinensis) juice (OJe) in diet-induced obese zebrafish. Methods: Adult zebrafish were divided into four diet groups: (i) normally fed (NF); (ii) overfed (OF); (iii) NF supplemented with OJe (5 mL/L in fish water; NF + OJe); and (iv) OF supplemented with OJe (OF + OJe). Each week, body weight (BW) and body mass index (BMI) were measured, and, at the end of the fifth week, euthanized zebrafish were processed for both microscopic evaluations and qPCR analyses. Results: In OF zebrafish, OJe significantly decreased both BW and BMI values and lowered the visceral adipose tissue, while it had little effect in the NF group. Moreover, it significantly reduced adipocyte cell size in both NF and OF groups in both visceral and subcutaneous adipose tissues, as well as their number in OF fish. Finally, OJe modulated some obesity-related genes, such as leptin A, ghrelin, orexin, pro-opiomelanocortin (POMC), and neuropeptide Y (NPY), in both gut and brain. Conclusion: This study adds new insights into the anti-obesity properties of orange juice and its flavonoids, suggesting their role as weight management agents through a lipolytic action linked to a restoration of metabolism-regulating gene expression.
Collapse
|
39
|
Stanojlovic M, Pallais JP, Kotz CM. Chemogenetic Modulation of Orexin Neurons Reverses Changes in Anxiety and Locomotor Activity in the A53T Mouse Model of Parkinson's Disease. Front Neurosci 2019; 13:702. [PMID: 31417337 PMCID: PMC6682689 DOI: 10.3389/fnins.2019.00702] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/21/2019] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PD symptomology is recognized as heterogeneous and in addition to motor function decline includes cognitive, mood, sleep, and metabolic disorders. Previous studies showed early reductions in anxiety and locomotion in the A53T mice model of PD. Since inflammation and astrogliosis are an integral part of PD pathology and impair proper neuronal function, we were keen to investigate if behavioral changes in A53T mice are accompanied by increased inflammation and astrogliosis in the hippocampus (Hipp) and motor cortex (mCtx) brain regions involved in the regulation of anxiety and locomotion, respectively. To test this, we used 3-, 5-, and 7-month-old A53T mice to examine anxiety-like behavior, locomotion, and expression of inflammation and astrogliosis markers in the Hipp and mCtx. Further, we examined the presence of alpha-synuclein accumulation in orexin neurons and orexin neuronal loss. The data show early reductions in anxiety-like behavior as well as increased locomotor activity, which was accompanied by inflammation and astrogliosis in the Hipp and mCtx. Due to the persistence of the orexin neuron population in A53T mice and the involvement of orexin in anxiety and locomotor regulation, we hypothesized that chemogenetic modulation of orexin neurons would reverse the observed reductions in anxiety-like behavior and the increases in locomotor activity in these animals. We showed that chemogenetic activation of orexin neurons in A53T mice restores anxiety-like behavior back to control levels without affecting locomotor activity, whereas the inhibition of orexin neurons reverses the elevated locomotor activity without any effects on anxiety-like behavior. This study exemplifies the complex role of orexin neurons in this model of PD and demonstrates the novel finding that changes in locomotor and anxiety-like behavior are accompanied by inflammation and astrogliosis. Together, these data suggest that the orexin system may play a significant role in early and late stages of PD.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Catherine M Kotz
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States.,Minneapolis VA Health Care System, Geriatric Research, Education and Clinical Center, Minneapolis, MN, United States
| |
Collapse
|
40
|
Stanojlovic M, Pallais Yllescas JP, Vijayakumar A, Kotz C. Early Sociability and Social Memory Impairment in the A53T Mouse Model of Parkinson's Disease Are Ameliorated by Chemogenetic Modulation of Orexin Neuron Activity. Mol Neurobiol 2019; 56:8435-8450. [PMID: 31250383 DOI: 10.1007/s12035-019-01682-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/14/2019] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is a multi-layered progressive neurodegenerative disease. Signature motor system impairments are accompanied by a variety of other symptoms such as mood, sleep, metabolic, and cognitive disorders. Interestingly, social cognition impairments can be observed from the earliest stages of the disease, prior to the onset of the motor symptoms. In this study, we investigated age-related reductions in sociability and social memory in the A53T mouse model of PD. Since inflammation and astrogliosis are an integral part of PD pathology and impair proper neuronal function, we examined astrogliosis and inflammation markers and parvalbumin expression in medial pre-frontal cortex (mPFC), part of the brain responsible for social cognition regulation. Finally, we used DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) for the stimulation and inhibition of orexin neuronal activity to modulate sociability and social memory in A53T mice. We observed that social cognition impairment in A53T mice is accompanied by an increase in astrogliosis and inflammation markers, in addition to loss of parvalbumin neurons and inhibitory pre-synaptic terminals in the mPFC. Moreover, DREADD-induced activation of orexin neurons restores social cognition in the A53T mouse model of PD. SIGNIFICANCE STATEMENT: Social cognition is severely affected in the early stages of Parkinson's disease. In this study, we identified the A53T mouse as a model of social cognitive impairment in PD. Observed alterations in sociability and social memory are accompanied by loss of parvalbumin positive neurons and loss of inhibitory input to mPFC. Stimulating orexin neurons using a chemogenetic approach (DREADDs) ameliorated social cognitive impairment. This study identifies a role for orexin neurons in social cognition in PD and suggests potential therapeutic targets for PD-related social cognition impairments.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| | | | - Aarthi Vijayakumar
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Catherine Kotz
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.,GRECC, Minneapolis VA Health Care System, Minneapolis, MN, USA
| |
Collapse
|
41
|
Stanojlovic M, Pallais Yllescas JP, Mavanji V, Kotz C. Chemogenetic activation of orexin/hypocretin neurons ameliorates aging-induced changes in behavior and energy expenditure. Am J Physiol Regul Integr Comp Physiol 2019; 316:R571-R583. [PMID: 30726119 PMCID: PMC6589608 DOI: 10.1152/ajpregu.00383.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023]
Abstract
Aging affects numerous physiological processes, as well as behavior. A large number of these processes are regulated, at least partially, by hypothalamic orexin neurons, and orexin tone may decrease with normal aging. In this study, we hypothesized that designer receptors exclusively activated by designer drugs (DREADD) stimulation of orexin neuronal activity will ameliorate the effect of aging on behavioral and metabolic alterations in young and middle-aged mice. DREADD targeting was achieved by stereotaxic injection of AAV vectors (AAV2-hSyn-DIO-hM3D(Gq)-mCherry) into the lateral hypothalamus of 5- and 12-mo old orexin-cre female mice and was confirmed by immunohistochemistry (IHC) analysis of orexin A and mCherry expression. After recovery, animals were subjected to a behavioral test battery consisting of the elevated plus maze (EPM), open field (OFT), and novel object recognition tests (NORT) to assess effects of aging on anxiety-like behavior, general locomotion, and working memory. A comprehensive laboratory animal monitoring system (CLAMS) was used to measure spontaneous physical activity (SPA) and energy expenditure (EE). The results indicate that activation of orexin neurons mitigates aging-induced reductions in anxiety-like behavior in middle-aged mice (P < 0.005) and increases locomotion in both young and middle-aged mice (P < 0.05). Activation of orexin neurons increases SPA (P < 0.01) and EE (P < 0.005) in middle-aged mice, restoring the levels to that observed in young animals. Results from this study identify orexin neurons as potential therapeutic targets for age-related impairments in cognitive and anxiety-related behavior, and energy balance.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota
| | | | - Vijaya Mavanji
- Minneapolis Veterans Affairs Health Care System, Geriatric Research Education and Clinical Center , Minneapolis, Minnesota
| | - Catherine Kotz
- Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota
- Minneapolis Veterans Affairs Health Care System, Geriatric Research Education and Clinical Center , Minneapolis, Minnesota
| |
Collapse
|
42
|
Pei H, Patterson CM, Sutton AK, Burnett KH, Myers MG, Olson DP. Lateral Hypothalamic Mc3R-Expressing Neurons Modulate Locomotor Activity, Energy Expenditure, and Adiposity in Male Mice. Endocrinology 2019; 160:343-358. [PMID: 30541071 PMCID: PMC6937456 DOI: 10.1210/en.2018-00747] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/04/2018] [Indexed: 02/05/2023]
Abstract
The central melanocortin system plays a crucial role in the control of energy balance. Although the decreased energy expenditure and increased adiposity of melanocortin-3 receptor (Mc3R)-null mice suggest the importance of Mc3R-regulated neurons in energy homeostasis, the roles for specific subsets of Mc3R neurons in energy balance have yet to be determined. Because the lateral hypothalamic area (LHA) contributes to the control of energy expenditure and feeding, we generated Mc3rcre mice to determine the roles of LHA Mc3R (Mc3RLHA) neurons in energy homeostasis. We found that Mc3RLHA neurons overlap extensively with LHA neuron markers that contribute to the control of energy balance (neurotensin, galanin, and leptin receptor) and project to brain areas involved in the control of feeding, locomotion, and energy expenditure, consistent with potential roles for Mc3RLHA neurons in these processes. Indeed, selective chemogenetic activation of Mc3RLHA neurons increased locomotor activity and augmented refeeding after a fast. Although the ablation of Mc3RLHA neurons did not alter food intake, mice lacking Mc3RLHA neurons displayed decreased energy expenditure and locomotor activity, along with increased body mass and adiposity. Thus, Mc3R neurons lie within LHA neurocircuitry that modulates locomotor activity and energy expenditure and contribute to energy balance control.
Collapse
Affiliation(s)
- Hongjuan Pei
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | | | - Amy K Sutton
- Molecular and Integrative Physiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Korri H Burnett
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | - Martin G Myers
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan
- Molecular and Integrative Physiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - David P Olson
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
- Molecular and Integrative Physiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
- Correspondence: David P. Olson, MD, PhD, University of Michigan, 1000 Wall Street, Brehm Tower 6329, Ann Arbor, Michigan 48105. E-mail:
| |
Collapse
|
43
|
Qualls-Creekmore E, Münzberg H. Modulation of Feeding and Associated Behaviors by Lateral Hypothalamic Circuits. Endocrinology 2018; 159:3631-3642. [PMID: 30215694 PMCID: PMC6195675 DOI: 10.1210/en.2018-00449] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022]
Abstract
Our ability to modulate and observe neuronal activity in defined neurons in freely moving animals has revolutionized neuroscience research in recent years. Findings in the lateral hypothalamus (LHA) highlighted the existence of many neuronal circuits that regulate distinct phenotypes of feeding behavior, emotional valence, and locomotor activity. Several of these neuronal circuits do not fit into a common model of neuronal integration and highlight the need to improve working models for complex behaviors. This review will specifically focus on recent literature that distinguishes LHA circuits based on their molecular and anatomical characteristics and studies their role in feeding, associated behaviors (e.g., arousal and locomotion), and emotional states (e.g., emotional valences).
Collapse
Affiliation(s)
- Emily Qualls-Creekmore
- Neurobiology of Nutrition and Metabolism, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Heike Münzberg
- Neurobiology of Nutrition and Metabolism, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| |
Collapse
|
44
|
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
|
45
|
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
|