1
|
Xing M, Li Y, Zhang Y, Zhou J, Ma D, Zhang M, Tang M, Ouyang T, Zhang F, Shi X, Sun J, Chen Z, Zhang WJ, Zhang S, Xie X. Paraventricular hypothalamic RUVBL2 neurons suppress appetite by enhancing excitatory synaptic transmission in distinct neurocircuits. Nat Commun 2024; 15:8939. [PMID: 39414808 PMCID: PMC11484884 DOI: 10.1038/s41467-024-53258-6] [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: 03/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The paraventricular hypothalamus (PVH) is crucial for food intake control, yet the presynaptic mechanisms underlying PVH neurons remain unclear. Here, we show that RUVBL2 in the PVH is significantly reduced during energy deficit, and knockout (KO) of PVH RUVBL2 results in hyperphagic obesity in mice. RUVBL2-expressing neurons in the PVH (PVHRUVBL2) exert the anorexigenic effect by projecting to the arcuate hypothalamus, the dorsomedial hypothalamus, and the parabrachial complex. We further demonstrate that PVHRUVBL2 neurons form the synaptic connections with POMC and AgRP neurons in the ARC. PVH RUVBL2 KO impairs the excitatory synaptic transmission by reducing presynaptic boutons and synaptic vesicles near active zone. Finally, RUVBL2 overexpression in the PVH suppresses food intake and protects against diet induced obesity. Together, this study demonstrates an essential role for PVH RUVBL2 in food intake control, and suggests that modulation of synaptic plasticity could be an effective way to curb appetite and obesity.
Collapse
Affiliation(s)
- Mingming Xing
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Juemou Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Danting Ma
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Mengqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Minglei Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ting Ouyang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Fumiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Weiping J Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
2
|
Münzberg H, Heymsfield SB, Berthoud HR, Morrison CD. History and future of leptin: Discovery, regulation and signaling. Metabolism 2024; 161:156026. [PMID: 39245434 DOI: 10.1016/j.metabol.2024.156026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
The cloning of leptin 30 years ago in 1994 was an important milestone in obesity research. Prior to the discovery of leptin, obesity was stigmatized as a condition caused by lack of character and self-control. Mutations in either leptin or its receptor were the first single gene mutations found to cause severe obesity, and it is now recognized that obesity is caused mostly by a dysregulation of central neuronal circuits. Since the discovery of the leptin-deficient obese mouse (ob/ob) the cloning of leptin (ob aka lep) and leptin receptor (db aka lepr) genes, we have learned much about leptin and its action in the central nervous system. The first hope that leptin would cure obesity was quickly dampened because humans with obesity have increased leptin levels and develop leptin resistance. Nevertheless, leptin target sites in the brain represent an excellent blueprint to understand how neuronal circuits control energy homeostasis. Our expanding understanding of leptin function, interconnection of leptin signaling with other systems and impact on distinct physiological functions continues to guide and improve the development of safe and effective interventions to treat metabolic illnesses. This review highlights past concepts and current emerging concepts of the hormone leptin, leptin receptor signaling pathways and central targets to mediate distinct physiological functions.
Collapse
Affiliation(s)
- Heike Münzberg
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America.
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Christopher D Morrison
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| |
Collapse
|
3
|
Srour N, Caron A, Michael NJ. Do POMC neurons have a sweet tooth for leptin? Special issue: Role of nutrients in nervous control of energy balance. Biochimie 2024; 223:179-187. [PMID: 36122808 DOI: 10.1016/j.biochi.2022.09.006] [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: 05/21/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022]
Abstract
Coordinated detection of changes in metabolic state by the nervous system is fundamental for survival. Hypothalamic pro-opiomelanocortin (POMC) neurons play a critical role in integrating metabolic signals, including leptin levels. They also coordinate adaptative responses and thus represent an important relay in the regulation of energy balance. Despite a plethora of work documenting the effects of individual hormones, nutrients, and neuropeptides on POMC neurons, the importance for crosstalk and additive effects between such signaling molecules is still underexplored. The ability of the metabolic state and the concentrations of nutrients, such as glucose, to influence leptin's effects on POMC neurons appears critical for understanding the function and complexity of this regulatory network. Here, we summarize the current knowledge on the effects of leptin on POMC neuron electrical excitability and discuss factors potentially contributing to variability in these effects, with a particular focus on the mouse models that have been developed and the importance of extracellular glucose levels. This review highlights the importance of the metabolic "environment" for determining hypothalamic neuronal responsiveness to metabolic cues and for determining the fundamental effects of leptin on the activity of hypothalamic POMC neurons.
Collapse
Affiliation(s)
- Nader Srour
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada; Montreal Diabetes Research Center, QC, Canada.
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
4
|
Ferrario CR, Münzberg-Gruening H, Rinaman L, Betley JN, Borgland SL, Dus M, Fadool DA, Medler KF, Morton GJ, Sandoval DA, de La Serre CB, Stanley SA, Townsend KL, Watts AG, Maruvada P, Cummings D, Cooke BM. Obesity- and diet-induced plasticity in systems that control eating and energy balance. Obesity (Silver Spring) 2024; 32:1425-1440. [PMID: 39010249 PMCID: PMC11269035 DOI: 10.1002/oby.24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 07/17/2024]
Abstract
In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.
Collapse
Grants
- P30 DK048520 NIDDK NIH HHS
- NSF1949989 National Science Foundation
- T32 DC000044 NIDCD NIH HHS
- R01 DK133464 NIDDK NIH HHS
- R01 DK089056 NIDDK NIH HHS
- R01 DK130246 NIDDK NIH HHS
- R01 DK124801 NIDDK NIH HHS
- R01 DK100685 NIDDK NIH HHS
- R01 DK124238 NIDDK NIH HHS
- R01 DK130875 NIDDK NIH HHS
- R01 DK125890 NIDDK NIH HHS
- Z99 DK999999 Intramural NIH HHS
- R01 DK124461 NIDDK NIH HHS
- K26 DK138368 NIDDK NIH HHS
- R01 DK121995 NIDDK NIH HHS
- R01 DK121531 NIDDK NIH HHS
- P30 DK089503 NIDDK NIH HHS
- P01 DK119130 NIDDK NIH HHS
- R01 DK118910 NIDDK NIH HHS
- R01 AT011683 NCCIH NIH HHS
- Reported research was supported by DK130246, DK092587, AT011683, MH059911, DK100685, DK119130, DK124801, DK133399, AG079877, DK133464, T32DC000044, F31DC016817, NSF1949989, DK089056, DK124238, DK138368, DK121995, DK125890, DK118910, DK121531, DK124461, DK130875; Canada Research Chair: 950-232211, CIHRFDN148473, CIHRPJT185886; USDA Predoctoral Fellowship; Endowment from the Robinson Family and Tallahassee Memorial Hospital; Department of Defense W81XWH-20-1-0345 and HT9425-23-1-0244; American Diabetes Association #1-17-ACE-31; W.M. Keck Foundation Award; National Science Foundation CAREER 1941822
- R01 DK133399 NIDDK NIH HHS
- HT9425-23-1-0244 Department of Defense
- R01 DK092587 NIDDK NIH HHS
- W81XWH-20-1-0345 Department of Defense
- 1941822 National Science Foundation
- R01 MH059911 NIMH NIH HHS
- F31 DC016817 NIDCD NIH HHS
- R01 AG079877 NIA NIH HHS
- P30 DK017047 NIDDK NIH HHS
Collapse
Affiliation(s)
- Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Heike Münzberg-Gruening
- Laboratory of Central Leptin Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Debra A Fadool
- Department of Biological Science, Program in Neuroscience, Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Kathryn F Medler
- School of Animal Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute at South Lake Union, Seattle, Washington, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Alan G Watts
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Padma Maruvada
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diana Cummings
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Bradley M Cooke
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Wang J, O'Reilly M, Cooper IA, Chehrehasa F, Moody H, Beecher K. Mapping GABAergic projections that mediate feeding. Neurosci Biobehav Rev 2024; 163:105743. [PMID: 38821151 DOI: 10.1016/j.neubiorev.2024.105743] [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: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Neuroscience offers important insights into the pathogenesis and treatment of obesity by investigating neural circuits underpinning appetite and feeding. Gamma-aminobutyric acid (GABA), one of the most abundant neurotransmitters in the brain, and its associated receptors represent an array of pharmacologically targetable mediators of appetite signalling. Targeting the GABAergic system is therefore an increasingly investigated approach to obesity treatment. However, the many GABAergic projections that control feeding have yet to be collectively analysed. This review provides a comprehensive analysis of the relationship between GABAergic signalling and appetite by examining both foundational studies and the results of newly emerging chemogenetic/optogenetic experiments. A current snapshot of these efforts to map GABAergic projections influencing appetite is provided, and potential avenues for further investigation are provided.
Collapse
Affiliation(s)
- Joshua Wang
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia.
| | - Max O'Reilly
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| | | | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Hayley Moody
- Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Kate Beecher
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| |
Collapse
|
6
|
Villa PA, Ruggiero-Ruff RE, Jamieson BB, Campbell RE, Coss D. Obesity Alters POMC and Kisspeptin Neuron Cross Talk Leading to Reduced Luteinizing Hormone in Male Mice. J Neurosci 2024; 44:e0222242024. [PMID: 38744532 PMCID: PMC11236585 DOI: 10.1523/jneurosci.0222-24.2024] [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: 02/01/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity is associated with hypogonadism in males, characterized by low testosterone and sperm number. Previous studies determined that these stem from dysregulation of hypothalamic circuitry that regulates reproduction, by unknown mechanisms. Herein, we used mice fed chronic high-fat diet, which mimics human obesity, to determine mechanisms of impairment at the level of the hypothalamus, in particular gonadotropin-releasing hormone (GnRH) neurons that regulate luteinizing hormone (LH), which then regulates testosterone. Consistent with obese humans, we demonstrated lower LH, and lower pulse frequency of LH secretion, but unchanged pituitary responsiveness to GnRH. LH pulse frequency is regulated by pulsatile GnRH secretion, which is controlled by kisspeptin. Peripheral and central kisspeptin injections, and DREADD-mediated activation of kisspeptin neurons, demonstrated that kisspeptin neurons were suppressed in obese mice. Thus, we investigated regulators of kisspeptin secretion. We determined that the LH response to NMDA was lower in obese mice, corresponding to fewer glutamate receptors in kisspeptin neurons, which may be critical for kisspeptin synchronization. Given that kisspeptin neurons also interact with anorexigenic POMC neurons, which are affected by obesity, we examined their cross talk, and determined that the LH response to either DREADD-mediated activation of POMC neurons or central injection of αMSH, a product of POMC, is abolished in obese mice. This was accompanied by diminished levels of αMSH receptor, MC4R, in kisspeptin neurons. Together, our studies determined that obesity leads to the downregulation of receptors that regulate kisspeptin neurons, which is associated with lower LH pulse frequency, leading to lower LH and hypogonadism.
Collapse
Affiliation(s)
- Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Bradley B Jamieson
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| |
Collapse
|
7
|
Frago LM, Gómez-Romero A, Collado-Pérez R, Argente J, Chowen JA. Synergism Between Hypothalamic Astrocytes and Neurons in Metabolic Control. Physiology (Bethesda) 2024; 39:0. [PMID: 38530221 DOI: 10.1152/physiol.00009.2024] [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: 02/01/2024] [Revised: 03/05/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024] Open
Abstract
Astrocytes are no longer considered as passive support cells. In the hypothalamus, these glial cells actively participate in the control of appetite, energy expenditure, and the processes leading to obesity and its secondary complications. Here we briefly review studies supporting this conclusion and the advances made in understanding the underlying mechanisms.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Gómez-Romero
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
8
|
Smith A, MacAulay B, Scheufen J, Hudak A, Abizaid A. Chronic Social Defeat Stress Increases Brain Permeability to Ghrelin in Male Mice. eNeuro 2024; 11:ENEURO.0093-24.2024. [PMID: 38937108 PMCID: PMC11253241 DOI: 10.1523/eneuro.0093-24.2024] [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: 03/04/2024] [Revised: 06/05/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
Ghrelin is a stomach-derived hormone that increases feeding and is elevated in response to chronic psychosocial stressors. The effects of ghrelin on feeding are mediated by the binding of ghrelin to the growth hormone secretagogue receptor (GHSR), a receptor located in hypothalamic and extrahypothalamic regions important for regulating food intake and metabolic rate. The ability of ghrelin to enter the brain, however, seems to be restricted to circumventricular organs like the median eminence and the brainstem area postrema, whereas ghrelin does not readily enter other GHSR-expressing regions like the ventral tegmental area (VTA). Interestingly, social stressors result in increased blood-brain barrier permeability, and this could therefore facilitate the entry of ghrelin into the brain. To investigate this, we exposed mice to social defeat stress for 21 d and then peripherally injected a Cy5-labelled biologically active ghrelin analog. The results demonstrate that chronically stressed mice exhibit higher Cy5-ghrelin fluorescence in several hypothalamic regions in addition to the ARC, including the hippocampus and midbrain. Furthermore, Cy5-ghrelin injections resulted in increased FOS expression in regions associated with the reward system in chronically stressed mice. Further histologic analyses identified a reduction in the branching of hypothalamic astrocytes in the ARC-median eminence junction, suggesting increased blood-brain barrier permeability. These data support the hypothesis that during metabolically challenging conditions like chronic stress, ghrelin may be more able to cross the blood-brain barrier and diffuse throughout the brain to target GHSR-expressing brain regions away from circumventricular organs.
Collapse
Affiliation(s)
- Andrea Smith
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Brenna MacAulay
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Jessica Scheufen
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Abagael Hudak
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
9
|
Wulff BS, Kuhre RE, Selvaraj M, Rehfeld JF, Niss K, Fels JJ, Anna S, Raun K, Gerstenberg MK. Improved leptin sensitivity and increased soluble leptin receptor concentrations may underlie the additive effects of combining PYY [, , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ] and exendin-4 on body weight lowering in diet-induced obese mice. Heliyon 2024; 10:e32009. [PMID: 39183855 PMCID: PMC11341243 DOI: 10.1016/j.heliyon.2024.e32009] [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: 09/18/2023] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 08/27/2024] Open
Abstract
Objective Co-treatment with long acting PYY and the GLP-1 receptor agonists has potential as an efficient obesity treatment. This study investigates whether the mechanisms behind additive reduction of food intake and weight loss depends on complementary effects in brain areas regulating food intake and if restoration of leptin sensitivity is involved. Methods Diet-induced obese (DIO) mice were co-treated with PYY(3-36) and exendin-4 (Ex4, GLP-1R agonist) for 14 days using minipumps. Leptin responsiveness was evaluated by measuring food intake and body weight after leptin injection, and gene expression profile was investigated in various of brain regions and liver. Results We show that weight loss associated with co-treatment of PYY(3-36) and Ex4 and Ex4 mono-treatment in DIO mice increased expression of several genes in area postrema (AP) known to be involved in appetite regulation and Cart, Pdyn, Bdnf and Klb were synergistically upregulated by the co-treatment. The upregulations were independent of weight loss, as shown by inclusion of a weight matched control. Moreover, PYY(3-36) and Ex4 co-treatment resulted in synergistically upregulated plasma concentrations of soluble leptin receptor (SLR) and improved sensitivity to exogenous leptin demonstrated by food intake lowering. Conclusion The study results suggest that synergistic upregulation of appetite-regulating genes in AP and improved leptin sensitivity are important mediators for the additive weight loss resulting from PYY and Ex4 co-treatment.
Collapse
Affiliation(s)
| | | | - Madhan Selvaraj
- Translational Research, Global Translation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Jens F. Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kristoffer Niss
- Biomarker Discovery, R&ED Digital Science and Innovation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Johannes J. Fels
- Research Bioanalysis, Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Secher Anna
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Kirsten Raun
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | | |
Collapse
|
10
|
Mazuecos L, Artigas-Jerónimo S, Pintado C, Gómez O, Rubio B, Arribas C, Andrés A, Villar M, Gallardo N. Central leptin signaling deficiency induced by leptin receptor antagonist leads to hypothalamic proteomic remodeling. Life Sci 2024; 346:122649. [PMID: 38626868 DOI: 10.1016/j.lfs.2024.122649] [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: 11/29/2023] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
AIMS Leptin irresponsiveness, which is often associated with obesity, can have significant impacts on the hypothalamic proteome of individuals, including those who are lean. While mounting evidence on leptin irresponsiveness has focused on obese individuals, understanding the early molecular and proteomic changes associated with deficient hypothalamic leptin signaling in lean individuals is essential for early intervention and prevention of metabolic disorders. Leptin receptor antagonists block the binding of leptin to its receptors, potentially reducing its effects and used in cases where excessive leptin activity might be harmful. MATERIALS AND METHODS In this work, we blocked the central actions of leptin in lean male adult Wistar rat by chronically administering intracerebroventricularly the superactive leptin receptor antagonist (SLA) (D23L/L39A/D40A/F41A) and investigated its impact on the hypothalamic proteome using label-free sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) for quantitative proteomics. KEY FINDINGS Our results show an accumulation of proteins involved in mRNA processing, mRNA stability, and translation in the hypothalamus of SLA-treated rats. Conversely, hypothalamic leptin signaling deficiency reduces the representation of proteins implicated in energy metabolism, neural circuitry, and neurotransmitter release. SIGNIFICANCE The alterations in the adult rat hypothalamic proteome contribute to dysregulate appetite, metabolism, and energy balance, which are key factors in the development and progression of obesity and related metabolic disorders. Additionally, using bioinformatic analysis, we identified a series of transcription factors that are potentially involved in the upstream regulatory mechanisms responsible for the observed signature.
Collapse
Affiliation(s)
- Lorena Mazuecos
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Sara Artigas-Jerónimo
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Cristina Pintado
- Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Avda. Carlos III s/n, 45071 Toledo, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Oscar Gómez
- Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Avda. Carlos III s/n, 45071 Toledo, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Blanca Rubio
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Carmen Arribas
- Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Avda. Carlos III s/n, 45071 Toledo, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Antonio Andrés
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Margarita Villar
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain.
| | - Nilda Gallardo
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain.
| |
Collapse
|
11
|
Cornejo MP, Fernandez G, Cabral A, Barrile F, Heredia F, García Romero G, Zubimendi Sampieri JP, Quelas JI, Cantel S, Fehrentz JA, Alonso A, Pla R, Ferran JL, Andreoli MF, De Francesco PN, Perelló M. GHSR in a Subset of GABA Neurons Controls Food Deprivation-Induced Hyperphagia in Male Mice. Endocrinology 2024; 165:bqae061. [PMID: 38815068 DOI: 10.1210/endocr/bqae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The growth hormone secretagogue receptor (GHSR), primarily known as the receptor for the hunger hormone ghrelin, potently controls food intake, yet the specific Ghsr-expressing cells mediating the orexigenic effects of this receptor remain incompletely characterized. Since Ghsr is expressed in gamma-aminobutyric acid (GABA)-producing neurons, we sought to investigate whether the selective expression of Ghsr in a subset of GABA neurons is sufficient to mediate GHSR's effects on feeding. First, we crossed mice that express a tamoxifen-dependent Cre recombinase in the subset of GABA neurons that express glutamic acid decarboxylase 2 (Gad2) enzyme (Gad2-CreER mice) with reporter mice, and found that ghrelin mainly targets a subset of Gad2-expressing neurons located in the hypothalamic arcuate nucleus (ARH) and that is predominantly segregated from Agouti-related protein (AgRP)-expressing neurons. Analysis of various single-cell RNA-sequencing datasets further corroborated that the primary subset of cells coexpressing Gad2 and Ghsr in the mouse brain are non-AgRP ARH neurons. Next, we crossed Gad2-CreER mice with reactivable GHSR-deficient mice to generate mice expressing Ghsr only in Gad2-expressing neurons (Gad2-GHSR mice). We found that ghrelin treatment induced the expression of the marker of transcriptional activation c-Fos in the ARH of Gad2-GHSR mice, yet failed to induce food intake. In contrast, food deprivation-induced refeeding was higher in Gad2-GHSR mice than in GHSR-deficient mice and similar to wild-type mice, suggesting that ghrelin-independent roles of GHSR in a subset of GABA neurons is sufficient for eliciting full compensatory hyperphagia in mice.
Collapse
Affiliation(s)
- María Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Gimena Fernandez
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Agustina Cabral
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Franco Barrile
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Florencia Heredia
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Guadalupe García Romero
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | | | | | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - Ramon Pla
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - María Florencia Andreoli
- Instituto de Desarrollo e Investigaciones Pediátricas (IDIP), HIAEP Sor María Ludovica de La Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala 751 24, Sweden
| | - Pablo Nicolas De Francesco
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala 751 24, Sweden
| |
Collapse
|
12
|
Collado-Perez R, Chamoso-Sánchez D, García A, Fernández-Alfonso MS, Jiménez-Hernáiz M, Canelles S, Argente J, Frago LM, Chowen JA. The differential effects of palmitic acid and oleic acid on the metabolic response of hypothalamic astrocytes from male and female mice. J Neurosci Res 2024; 102:e25339. [PMID: 38741550 DOI: 10.1002/jnr.25339] [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: 11/28/2023] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
Diets rich in saturated fats are more detrimental to health than those containing mono- or unsaturated fats. Fatty acids are an important source of energy, but they also relay information regarding nutritional status to hypothalamic metabolic circuits and when in excess can be detrimental to these circuits. Astrocytes are the main site of central fatty acid β-oxidation, and hypothalamic astrocytes participate in energy homeostasis, in part by modulating hormonal and nutritional signals reaching metabolic neurons, as well as in the inflammatory response to high-fat diets. Thus, we hypothesized that how hypothalamic astrocytes process-specific fatty acids participates in determining the differential metabolic response and that this is sex dependent as males and females respond differently to high-fat diets. Male and female primary hypothalamic astrocyte cultures were treated with oleic acid (OA) or palmitic acid (PA) for 24 h, and an untargeted metabolomics study was performed. A clear predictive model for PA exposure was obtained, while the metabolome after OA exposure was not different from controls. The observed modifications in metabolites, as well as the expression levels of key metabolic enzymes, indicate a reduction in the activity of the Krebs and glutamate/glutamine cycles in response to PA. In addition, there were specific differences between the response of astrocytes from male and female mice, as well as between hypothalamic and cerebral cortical astrocytes. Thus, the response of hypothalamic astrocytes to specific fatty acids could result in differential impacts on surrounding metabolic neurons and resulting in varied systemic metabolic outcomes.
Collapse
Affiliation(s)
- Roberto Collado-Perez
- Department of Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Chamoso-Sánchez
- Center for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Antonia García
- Center for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | | | - Maria Jiménez-Hernáiz
- Department of Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
13
|
Başer Ö, Yavuz Y, Özen DÖ, Özgün HB, Ağuş S, Civaş CC, Atasoy D, Yılmaz B. Effects of chronic high fat diet on mediobasal hypothalamic satiety neuron function in POMC-Cre mice. Mol Metab 2024; 82:101904. [PMID: 38395148 PMCID: PMC10910127 DOI: 10.1016/j.molmet.2024.101904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE The prevalence of obesity has increased over the past three decades. Proopiomelanocortin (POMC) neurons in the hypothalamic arcuate nucleus (ARC) play a vital role in induction of satiety. Chronic consumption of high-fat diet is known to reduce hypothalamic neuronal sensitivity to hormones like leptin, thus contributing to the development and persistence of obesity. The functional and morphological effects of a high-calorie diet on POMC neurons and how these effects contribute to the development and maintenance of the obese phenotype are not fully understood. For this purpose, POMC-Cre transgenic mice model was exposed to high-fat diet (HFD) and at the end of a 3- and 6-month period, electrophysiological and morphological changes, and the role of POMC neurons in homeostatic nutrition and their response to leptin were thoroughly investigated. METHODS Effects of HFD on POMC-satiety neurons in transgenic mice models exposed to chronic high-fat diet were investigated using electrophysiological (patch-clamp), chemogenetic and Cre recombinase advanced technological methods. Leptin, glucose and lipid profiles were determined and analyzed. RESULTS In mice exposed to a high-fat diet for 6 months, no significant changes in POMC dendritic spine number or projection density from POMC neurons to the paraventricular hypothalamus (PVN), lateral hypothalamus (LH), and bed nucleus stria terminalis (BNST) were observed. It was revealed that leptin hormone did not change the electrophysiological activities of POMC neurons in mice fed with HFD for 6 months. In addition, chemogenetic stimulation of POMC neurons increased HFD consumption. In the 3-month HFD-fed group, POMC activation induced an orexigenic response in mice, whereas switching to a standard diet was found to abolish orexigenic behavior in POMC mice. CONCLUSIONS Chronic high fat consumption disrupts the regulation of POMC neuron activation by leptin. Altered POMC neuron activation abolished the neuron's characteristic behavioral anorexigenic response. Change in nutritional content contributes to the reorganization of developing maladaptations.
Collapse
Affiliation(s)
- Özge Başer
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Yavuz Yavuz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Deniz Öykü Özen
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Hüseyin Buğra Özgün
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Sami Ağuş
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Cihan Civan Civaş
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Deniz Atasoy
- University of Iowa, Carver College of Medicine, Department of Neuroscience and Pharmacology, Iowa City, USA
| | - Bayram Yılmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye; Izmir Biomedicine and Genome Center, Izmir, Türkiye.
| |
Collapse
|
14
|
Ameroso D, Rios M. Synaptic plasticity and the role of astrocytes in central metabolic circuits. WIREs Mech Dis 2024; 16:e1632. [PMID: 37833830 PMCID: PMC10842964 DOI: 10.1002/wsbm.1632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023]
Abstract
Neural circuits in the brain, primarily in the hypothalamus, are paramount to the homeostatic control of feeding and energy utilization. They integrate hunger, satiety, and body adiposity cues from the periphery and mediate the appropriate behavioral and physiological responses to satisfy the energy demands of the animal. Notably, perturbations in central homeostatic circuits have been linked to the etiology of excessive feeding and obesity. Considering the ever-changing energy requirements of the animal and required adaptations, it is not surprising that brain-feeding circuits remain plastic in adulthood and are subject to changes in synaptic strength as a consequence of nutritional status. Indeed, synapse density, probability of presynaptic transmitter release, and postsynaptic responses in hypothalamic energy balance centers are tailored to behavioral and physiological responses required to sustain survival. Mounting evidence supports key roles of astrocytes facilitating some of this plasticity. Here we discuss these synaptic plasticity mechanisms and the emerging roles of astrocytes influencing energy and glucose balance control in health and disease. This article is categorized under: Cancer > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
15
|
Mata-Pacheco V, Hernandez J, Varma N, Xu J, Sayers S, Le N, Wagner EJ. Dynamic, sex- and diet-specific pleiotropism in the PAC1 receptor-mediated regulation of arcuate proopiomelanocortin and Neuropeptide Y/Agouti related peptide neuronal excitability by anorexigenic ventromedial nucleus PACAP neurons. J Neuroendocrinol 2024; 36:e13357. [PMID: 38056947 DOI: 10.1111/jne.13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
This study furthers the investigation of how pituitary adenylate cyclase activating polypeptide (PACAP) and the PAC1 receptor (PAC1R) regulate the homeostatic energy balance circuitry. We hypothesized that apoptotic ablation of PACAP neurones in the hypothalamic ventromedial nucleus (VMN) would affect both energy intake and energy expenditure. We also hypothesized that selective PAC1R knockdown would impair the PACAP-induced excitation in anorexigenic proopiomelanocortin (POMC) neurones and inhibition of orexigenic neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurones in the hypothalamic arcuate nucleus (ARC). The results show CASPASE-3-induced ablation of VMN PACAP neurones leads to increased energy intake and meal frequency as well as decreased energy expenditure in lean animals. The effects were more robust in obese males, whereas we saw the opposite effects in obese females. We then utilized visualized whole-cell patch clamp recordings in hypothalamic slices. PAC1R knockdown in POMC neurones diminishes the PACAP-induced depolarization, increase in firing, decreases in energy intake and meal size, as well as increases in CO2 production and O2 consumption. Similarly, the lack of expression of the PAC1R in NPY/AgRP neurones greatly attenuates the PACAP-induced hyperpolarization, suppression of firing, decreases in energy intake and meal frequency, as well as increases in energy expenditure. The PACAP response in NPY/AgRP neurones switched from predominantly inhibitory to excitatory in fasted animals. Finally, the anorexigenic effect of PACAP was potentiated when oestradiol was injected into the ARC in ovariectomized females. This study demonstrates the critical role of anorexigenic VMN PACAP neurones and the PAC1R in exciting POMC and inhibiting NPY/AgRP neurons to control homeostatic feeding.
Collapse
Affiliation(s)
- Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Jennifer Hernandez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Nandini Varma
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Jenny Xu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Sarah Sayers
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Nikki Le
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Edward J Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
16
|
Herb BR, Glover HJ, Bhaduri A, Colantuoni C, Bale TL, Siletti K, Hodge R, Lein E, Kriegstein AR, Doege CA, Ament SA. Single-cell genomics reveals region-specific developmental trajectories underlying neuronal diversity in the human hypothalamus. SCIENCE ADVANCES 2023; 9:eadf6251. [PMID: 37939194 PMCID: PMC10631741 DOI: 10.1126/sciadv.adf6251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
The development and diversity of neuronal subtypes in the human hypothalamus has been insufficiently characterized. To address this, we integrated transcriptomic data from 241,096 cells (126,840 newly generated) in the prenatal and adult human hypothalamus to reveal a temporal trajectory from proliferative stem cell populations to mature hypothalamic cell types. Iterative clustering of the adult neurons identified 108 robust transcriptionally distinct neuronal subtypes representing 10 hypothalamic nuclei. Pseudotime trajectories provided insights into the genes driving formation of these nuclei. Comparisons to single-cell transcriptomic data from the mouse hypothalamus suggested extensive conservation of neuronal subtypes despite certain differences in species-enriched gene expression. The uniqueness of hypothalamic neuronal lineages was examined developmentally by comparing excitatory lineages present in cortex and inhibitory lineages in ganglionic eminence, revealing both distinct and shared drivers of neuronal maturation across the human forebrain. These results provide a comprehensive transcriptomic view of human hypothalamus development through gestation and adulthood at cellular resolution.
Collapse
Affiliation(s)
- Brian R. Herb
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hannah J. Glover
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Carlo Colantuoni
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tracy L. Bale
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Hodge
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Arnold R. Kriegstein
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seth A. Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Fan S, Guo W, Xiao D, Guan M, Liao T, Peng S, Feng A, Wang Z, Yin H, Li M, Chen J, Xiong W. Microbiota-gut-brain axis drives overeating disorders. Cell Metab 2023; 35:2011-2027.e7. [PMID: 37794596 DOI: 10.1016/j.cmet.2023.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/10/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Overeating disorders (ODs), usually stemming from dieting history and stress, remain a pervasive issue in contemporary society, with the pathological mechanisms largely unresolved. Here, we show that alterations in intestinal microbiota are responsible for the excessive intake of palatable foods in OD mice and patients with bulimia nervosa (BN). Stress combined with a history of dieting causes significant changes in the microbiota and the intestinal metabolism, which disinhibit the vagus nerve terminals in the gut and thereby lead to a subsequent hyperactivation of the gut-brain axis passing through the vagus, the solitary tract nucleus, and the paraventricular nucleus of the thalamus. The transplantation of a probiotic Faecalibacterium prausnitzii or dietary supplement of key metabolites restores the activity of the gut-to-brain pathway and thereby alleviates the OD symptoms. Thus, our study delineates how the microbiota-gut-brain axis mediates energy balance, unveils the underlying pathogenesis of the OD, and provides potential therapeutic strategies.
Collapse
Affiliation(s)
- Sijia Fan
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Weiwei Guo
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Dan Xiao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Mengyuan Guan
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Tiepeng Liao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Sufang Peng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Airong Feng
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Ziyi Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Hao Yin
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Min Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230026, China.
| | - Jue Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Wei Xiong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; Anhui Province Key Laboratory of Biomedical Aging Research, Hefei 230026, China.
| |
Collapse
|
18
|
Wang X, Wu X, Wu H, Xiao H, Hao S, Wang B, Li C, Bleymehl K, Kauschke SG, Mack V, Ferger B, Klein H, Zheng R, Duan S, Wang H. Neural adaption in midbrain GABAergic cells contributes to high-fat diet-induced obesity. SCIENCE ADVANCES 2023; 9:eadh2884. [PMID: 37910621 PMCID: PMC10619925 DOI: 10.1126/sciadv.adh2884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023]
Abstract
Overeating disorders largely contribute to worldwide incidences of obesity. Available treatments are limited. Here, we discovered that long-term chemogenetic activation of ventrolateral periaqueductal gray (vlPAG) GABAergic cells rescue obesity of high-fat diet-induced obesity (DIO) mice. This was associated with the recovery of enhanced mIPSCs, decreased food intake, increased energy expenditure, and inguinal white adipose tissue (iWAT) browning. In vivo calcium imaging confirmed vlPAG GABAergic suppression for DIO mice, with corresponding reduction in intrinsic excitability. Single-nucleus RNA sequencing identified transcriptional expression changes in GABAergic cell subtypes in DIO mice, highlighting Cacna2d1 as of potential importance. Overexpressing CACNA2D1 in vlPAG GABAergic cells of DIO mice rescued enhanced mIPSCs and calcium response, reversed obesity, and therefore presented here as a potential target for obesity treatment.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaotong Wu
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Wu
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang 310058, China
| | - Hanyang Xiao
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Sijia Hao
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100091, China
| | - Chen Li
- Department of Human Genetics and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Katherin Bleymehl
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Stefan G. Kauschke
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Volker Mack
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Boris Ferger
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Holger Klein
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, 88397, Germany
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100091, China
| | - Shumin Duan
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Wang
- Department of Neurosurgery of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Lingang Laboratory, Shanghai 200031, China
| |
Collapse
|
19
|
Liu Z, Xiao T, Liu H. Leptin signaling and its central role in energy homeostasis. Front Neurosci 2023; 17:1238528. [PMID: 38027481 PMCID: PMC10644276 DOI: 10.3389/fnins.2023.1238528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Leptin plays a critical role in regulating appetite, energy expenditure and body weight, making it a key factor in maintaining a healthy balance. Despite numerous efforts to develop therapeutic interventions targeting leptin signaling, their effectiveness has been limited, underscoring the importance of gaining a better understanding of the mechanisms through which leptin exerts its functions. While the hypothalamus is widely recognized as the primary site responsible for the appetite-suppressing and weight-reducing effects of leptin, other brain regions have also been increasingly investigated for their involvement in mediating leptin's action. In this review, we summarize leptin signaling pathways and the neural networks that mediate the effects of leptin, with a specific emphasis on energy homeostasis.
Collapse
Affiliation(s)
- Zhaoxun Liu
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Xiao
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hailan Liu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
20
|
Srour N, Lavoie O, Khouma A, Minbashi Moeini M, Plamondon J, Kinkead R, Michael NJ, Caron A. Electrophysiological Comparison of Definitive Pro-opiomelanocortin Neurons in the Arcuate Nucleus and the Retrochiasmatic Area of Male and Female Mice. Neuroscience 2023; 530:95-107. [PMID: 37619768 DOI: 10.1016/j.neuroscience.2023.08.024] [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: 04/18/2023] [Revised: 07/28/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus (ARC) are considered a major site of leptin action. Due to increasing evidence that POMC neurons are highly heterogeneous and indications that the conventional molecular tools to study their functions have important limitations, a reassessment of leptin's effects on definitive POMC neurons is needed. POMC neurons are also expressed in the retrochiasmatic area (RCA), where their function is poorly understood. Furthermore, the response of POMC neurons to leptin in females is largely unknown. Therefore, the present study aimed to determine the differences in leptin responsiveness of POMC neurons in the ARC and the RCA using a mouse model allowing adult-inducible fluorescent labeling. We performed whole-cell patch clamp electrophysiology on 154 POMC neurons from male and female mice. We confirmed and extended the model by which leptin depolarizes POMC neurons, in both the ARC and the RCA. Furthermore, we characterized the electrophysiological properties of an underappreciated subpopulation representing ∼10% of hypothalamic POMC neurons that are inhibited by leptin. We also provide evidence that sex does not appear to be a major determinant of basal properties and leptin responsiveness of POMC neurons, but that females are overall less responsive to leptin compared to males.
Collapse
Affiliation(s)
- Nader Srour
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada; Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Olivier Lavoie
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada; Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Axelle Khouma
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada; Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Moein Minbashi Moeini
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada; Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | | | - Richard Kinkead
- Quebec Heart and Lung Institute, Quebec City, QC, Canada; Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Natalie J Michael
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada; Quebec Heart and Lung Institute, Quebec City, QC, Canada.
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada; Quebec Heart and Lung Institute, Quebec City, QC, Canada.
| |
Collapse
|
21
|
Tan R, Hu X, Wang X, Sun M, Cai Z, Zhang Z, Fu Y, Chen X, An J, Lu H. Leptin Promotes the Proliferation and Neuronal Differentiation of Neural Stem Cells through the Cooperative Action of MAPK/ERK1/2, JAK2/STAT3 and PI3K/AKT Signaling Pathways. Int J Mol Sci 2023; 24:15151. [PMID: 37894835 PMCID: PMC10606644 DOI: 10.3390/ijms242015151] [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: 09/05/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The potential of neural stem cells (NSCs) for neurological disorders the treatment has relied in large part upon identifying the NSCs fate decision. The hormone leptin has been reported to be a crucial regulator of brain development, able to influence the glial and neural development, yet, the underlying mechanism of leptin acting on NSCs' biological characteristics is still poorly understood. This study aims to investigate the role of leptin in the biological properties of NSCs. In this study, we investigate the possibility that leptin may regulate the NSCs' fate decision, which may promote the proliferation and neuronal differentiation of NSCs and thus act positively in neurological disorders. NSCs from the embryonic cerebral cortex were used in this study. We used CCK-8 assay, ki67 immunostaining, and FACS analysis to confirm that 25-100 ng/mL leptin promotes the proliferation of NSCs in a concentration-dependent pattern. This change was accompanied by the upregulation of p-AKT and p-ERK1/2, which are the classical downstream signaling pathways of leptin receptors b (LepRb). Inhibition of PI3K/AKT or MAPK/ERK signaling pathways both abolished the effect of leptin-induced proliferation. Moreover, leptin also enhanced the directed neuronal differentiation of NSCs. A blockade of the PI3K/AKT pathway reversed leptin-stimulated neurogenesis, while a blockade of JAK2/STAT3 had no effect on it. Taken together, our results support a role for leptin in regulating the fate of NSCs differentiation and promoting NSCs proliferation, which could be a promising approach for brain repair via regulating the biological characteristics of NSCs.
Collapse
Affiliation(s)
- Ruolan Tan
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xiaoxuan Hu
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xinyi Wang
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Meiqi Sun
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Zhenlu Cai
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Zixuan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yali Fu
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xinlin Chen
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Jing An
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Haixia Lu
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| |
Collapse
|
22
|
Abstract
Eating behaviours are determined by the integration of interoceptive and environmental inputs. During pregnancy, numerous physiological adaptations take place in the maternal organism to provide an adequate environment for embryonic growth. Among them, whole-body physiological remodelling directly influences eating patterns, commonly causing notable taste perception alterations, food aversions and cravings. Recurrent food cravings for and compulsive eating of highly palatable food can contribute to the development and maintenance of gestational overweight and obesity with potential adverse health consequences for the offspring. Although much is known about how maternal eating habits influence offspring health, the mechanisms that underlie changes in taste perception and food preference during pregnancy (which guide and promote feeding) are only just starting to be elucidated. Given the limited and diffuse understanding of the neurobiology of gestational eating patterns, the aim of this Review is to compile, integrate and discuss the research conducted on this topic in both experimental models and humans. This article sheds light on the mechanisms that drive changes in female feeding behaviours during distinct physiological states. Understanding these processes is crucial to improve gestational parent health and decrease the burden of metabolic and food-related diseases in future generations.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
23
|
Brüning JC, Fenselau H. Integrative neurocircuits that control metabolism and food intake. Science 2023; 381:eabl7398. [PMID: 37769095 DOI: 10.1126/science.abl7398] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023]
Abstract
Systemic metabolism has to be constantly adjusted to the variance of food intake and even be prepared for anticipated changes in nutrient availability. Therefore, the brain integrates multiple homeostatic signals with numerous cues that predict future deviations in energy supply. Recently, our understanding of the neural pathways underlying these regulatory principles-as well as their convergence in the hypothalamus as the key coordinator of food intake, energy expenditure, and glucose metabolism-have been revealed. These advances have changed our view of brain-dependent control of metabolic physiology. In this Review, we discuss new concepts about how alterations in these pathways contribute to the development of prevalent metabolic diseases such as obesity and type 2 diabetes mellitus and how this emerging knowledge may provide new targets for their treatment.
Collapse
Affiliation(s)
- Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Henning Fenselau
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Research Group Synaptic Transmission in Energy Homeostasis, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| |
Collapse
|
24
|
Huber K, Szerenos E, Lewandowski D, Toczylowski K, Sulik A. The Role of Adipokines in the Pathologies of the Central Nervous System. Int J Mol Sci 2023; 24:14684. [PMID: 37834128 PMCID: PMC10572192 DOI: 10.3390/ijms241914684] [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: 08/31/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Adipokines are protein hormones secreted by adipose tissue in response to disruptions in physiological homeostasis within the body's systems. The regulatory functions of adipokines within the central nervous system (CNS) are multifaceted and intricate, and they have been identified in a number of pathologies. Therefore, specific adipokines have the potential to be used as biomarkers for screening purposes in neurological dysfunctions. The systematic review presented herein focuses on the analysis of the functions of various adipokines in the pathogenesis of CNS diseases. Thirteen proteins were selected for analysis through scientific databases. It was found that these proteins can be identified within the cerebrospinal fluid either by their ability to modify their molecular complex and cross the blood-brain barrier or by being endogenously produced within the CNS itself. As a result, this can correlate with their measurability during pathological processes, including Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, depression, or brain tumors.
Collapse
Affiliation(s)
| | | | | | - Kacper Toczylowski
- Department of Pediatric Infectious Diseases, Medical University of Bialystok, Waszyngtona 17, 15-274 Bialystok, Poland
| | | |
Collapse
|
25
|
Mansano NDS, Vieira HR, Araujo-Lopes R, Szawka RE, Donato J, Frazao R. Fasting Modulates GABAergic Synaptic Transmission to Arcuate Kisspeptin Neurons in Female Mice. Endocrinology 2023; 164:bqad150. [PMID: 37793082 DOI: 10.1210/endocr/bqad150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/06/2023]
Abstract
It is well-established that the hypothalamic-pituitary-gonadal (HPG) axis is suppressed due to negative energy balance. However, less information is available on whether kisspeptin neuronal activity contributes to fasting-induced responses. In the present study, female and male mice were fasted for 24 hours or provided food ad libitum (fed group) to determine whether acute fasting is sufficient to modulate kisspeptin neuronal activity. In female mice, fasting attenuated luteinizing hormone (LH) and prolactin (PRL) serum levels and increased follicle-stimulating hormone levels compared with the fed group. In contrast, fasting did not affect gonadotropin or PRL secretion in male mice. By measuring genes related to LH pulse generation in micropunches obtained from the arcuate nucleus of the hypothalamus (ARH), we observed that fasting reduced Kiss1 mRNA levels in female and male mice. In contrast, Pdyn expression was upregulated only in fasted female mice, whereas no changes in the Tac2 mRNA levels were observed in both sexes. Interestingly, the frequency and amplitude of the GABAergic postsynaptic currents recorded from ARH kisspeptin neurons (ARHKisspeptin) were reduced in 24-hour fasted female mice but not in males. Additionally, neuropeptide Y induced a hyperpolarization in the resting membrane potential of ARHKisspeptin neurons of fed female mice but not in males. Thus, the response of ARHKisspeptin neurons to fasting is sexually dependent with a female bias, associated with changes in gonadotropins and PRL secretion. Our findings suggest that GABAergic transmission to ARHKisspeptin neurons modulates the activity of the HPG axis during situations of negative energy balance.
Collapse
Affiliation(s)
- Naira da Silva Mansano
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| | - Henrique Rodrigues Vieira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| | - Roberta Araujo-Lopes
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Belo Horizonte, MG 31270-901, Brazil
| | - Raphael Escorsim Szawka
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Belo Horizonte, MG 31270-901, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, São Paulo, SP 05508-000, Brazil
| | - Renata Frazao
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
26
|
Molière S, Jaulin A, Tomasetto CL, Dali-Youcef N. Roles of Matrix Metalloproteinases and Their Natural Inhibitors in Metabolism: Insights into Health and Disease. Int J Mol Sci 2023; 24:10649. [PMID: 37445827 DOI: 10.3390/ijms241310649] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-activated peptidases that can be classified into six major classes, including gelatinases, collagenases, stromelysins, matrilysins, membrane type metalloproteinases, and other unclassified MMPs. The activity of MMPs is regulated by natural inhibitors called tissue inhibitors of metalloproteinases (TIMPs). MMPs are involved in a wide range of biological processes, both in normal physiological conditions and pathological states. While some of these functions occur during development, others occur in postnatal life. Although the roles of several MMPs have been extensively studied in cancer and inflammation, their function in metabolism and metabolic diseases have only recently begun to be uncovered, particularly over the last two decades. This review aims to summarize the current knowledge regarding the metabolic roles of metalloproteinases in physiology, with a strong emphasis on adipose tissue homeostasis, and to highlight the consequences of impaired or exacerbated MMP actions in the development of metabolic disorders such as obesity, fatty liver disease, and type 2 diabetes.
Collapse
Affiliation(s)
- Sébastien Molière
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- Department of Radiology, Strasbourg University Hospital, Hôpital de Hautepierre, Avenue Molière, 67200 Strasbourg, France
- Breast and Thyroid Imaging Unit, ICANS-Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Amélie Jaulin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Catherine-Laure Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
| | - Nassim Dali-Youcef
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- Laboratoire de Biochimie et Biologie Moléculaire, Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, 67000 Strasbourg, France
| |
Collapse
|
27
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in VTA dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541104. [PMID: 37293057 PMCID: PMC10245666 DOI: 10.1101/2023.05.17.541104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTA DA ) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear if the same or different VTA DA neurons encode these different stimuli. To address this question, we performed 2-photon calcium imaging in mice presented with food and conspecifics, and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that modifying motivation for one stimulus affects responses to both stimuli. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone related genes in individual VTA DA neurons. Taken together, our functional and transcriptional data suggest overlapping VTA DA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Adelaide R. Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Nirja Oak
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Stephan Y. Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Catherine J. Peña
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ilana B. Witten
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| |
Collapse
|
28
|
Grzelka K, Wilhelms H, Dodt S, Dreisow ML, Madara JC, Walker SJ, Wu C, Wang D, Lowell BB, Fenselau H. A synaptic amplifier of hunger for regaining body weight in the hypothalamus. Cell Metab 2023; 35:770-785.e5. [PMID: 36965483 PMCID: PMC10160008 DOI: 10.1016/j.cmet.2023.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 12/15/2022] [Accepted: 03/01/2023] [Indexed: 03/27/2023]
Abstract
Restricting caloric intake effectively reduces body weight, but most dieters fail long-term adherence to caloric deficit and eventually regain lost weight. Hypothalamic circuits that control hunger drive critically determine body weight; yet, how weight loss sculpts these circuits to motivate food consumption until lost weight is regained remains unclear. Here, we probe the contribution of synaptic plasticity in discrete excitatory afferents on hunger-promoting AgRP neurons. We reveal a crucial role for activity-dependent, remarkably long-lasting amplification of synaptic activity originating from paraventricular hypothalamus thyrotropin-releasing (PVHTRH) neurons in long-term body weight control. Silencing PVHTRH neurons inhibits the potentiation of excitatory input to AgRP neurons and diminishes concomitant regain of lost weight. Brief stimulation of the pathway is sufficient to enduringly potentiate this glutamatergic hunger synapse and triggers an NMDAR-dependent gaining of body weight that enduringly persists. Identification of this activity-dependent synaptic amplifier provides a previously unrecognized target to combat regain of lost weight.
Collapse
Affiliation(s)
- Katarzyna Grzelka
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Hannah Wilhelms
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Stephan Dodt
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Marie-Luise Dreisow
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Samuel J Walker
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chen Wu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daqing Wang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA.
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
29
|
Joye DAM, Rohr KE, Suenkens K, Wuorinen A, Inda T, Arzbecker M, Mueller E, Huber A, Pancholi H, Blackmore MG, Carmona-Alcocer V, Evans JA. Somatostatin regulates central clock function and circadian responses to light. Proc Natl Acad Sci U S A 2023; 120:e2216820120. [PMID: 37098068 PMCID: PMC10160998 DOI: 10.1073/pnas.2216820120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Daily and annual changes in light are processed by central clock circuits that control the timing of behavior and physiology. The suprachiasmatic nucleus (SCN) in the anterior hypothalamus processes daily photic inputs and encodes changes in day length (i.e., photoperiod), but the SCN circuits that regulate circadian and photoperiodic responses to light remain unclear. Somatostatin (SST) expression in the hypothalamus is modulated by photoperiod, but the role of SST in SCN responses to light has not been examined. Our results indicate that SST signaling regulates daily rhythms in behavior and SCN function in a manner influenced by sex. First, we use cell-fate mapping to provide evidence that SST in the SCN is regulated by light via de novo Sst activation. Next, we demonstrate that Sst -/- mice display enhanced circadian responses to light, with increased behavioral plasticity to photoperiod, jetlag, and constant light conditions. Notably, lack of Sst -/- eliminated sex differences in photic responses due to increased plasticity in males, suggesting that SST interacts with clock circuits that process light differently in each sex. Sst -/- mice also displayed an increase in the number of retinorecipient neurons in the SCN core, which express a type of SST receptor capable of resetting the molecular clock. Last, we show that lack of SST signaling modulates central clock function by influencing SCN photoperiodic encoding, network after-effects, and intercellular synchrony in a sex-specific manner. Collectively, these results provide insight into peptide signaling mechanisms that regulate central clock function and its response to light.
Collapse
Affiliation(s)
- Deborah A. M. Joye
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Kayla E. Rohr
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Kimberlee Suenkens
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Alissa Wuorinen
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Thomas Inda
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Madeline Arzbecker
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Emma Mueller
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Alec Huber
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Harshida Pancholi
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | | | | | - Jennifer A. Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| |
Collapse
|
30
|
McCormack JC, Roberts R, Garratt M, Wang T, Hayes J, Peng M. Longitudinal study of energy, neurosensory and eating responses durinG pregnancY (ENERGY cohort): A study protocol. Clin Nutr ESPEN 2023; 54:271-276. [PMID: 36963873 DOI: 10.1016/j.clnesp.2023.01.033] [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: 07/03/2022] [Revised: 01/08/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Physiological changes that occur during pregnancy can have long-term impacts on metabolism and neurosensory responses to food, which can impact nutrition and health outcomes. The ENERGY cohort is a longitudinal study that aims to capitalizes on pregnancy as a natural model of metabolic reprogramming in order to understand the neurosensory mechanisms underpinning links between metabolism and dietary behaviour. The study objectives are to test for multi-sensory shifts during pregnancy, and the effect of sensory changes on dietary choices and bodyweights, and to identify neurosensory mechanisms that determine macronutrient selection before and after pregnancy. METHODS A longitudinal cohort study involving 130 pregravid women planning to conceive with the next 12-months and 65 pregravid women with no short-term plans to conceive. Participants will be recruited from Dunedin and Auckland, New Zealand. The study will test for changes in diet, neurosensory outcomes, and metabolism across the reproductive cycle, from pre-pregnancy to 1-year post-pregnancy. Data will be collected at six timepoint throughout the pregnancy which will occur approximately every 3 months. The primary response variables will be changes in supra-threshold sensitivity across modalities, dietary intake, and metabolism between pre-pregnancy and post-pregnancy. Longitudinal data analysis will use linear mixed models to assess changes in the response outcomes over time adjusted for age, ethnicity, and socioeconomic status. DISCUSSION Understanding the relationship between metabolism, sensory processing, and macronutrient preferences will provide crucial insights into diet-related health issues, including obesity. This study will lead to the formation of a prospective research cohort that is unique to Aotearoa New Zealand, and will develop multidisciplinary skills that are increasingly necessary to addressing the obesity epidemic.
Collapse
Affiliation(s)
- Jessica C McCormack
- Sensory Neurosensory and Nutrtion Laboratory, Department of Food Science, University of Otago, Dunedin, New Zealand
| | - Reece Roberts
- School of Psychology, University of Auckland, Auckland, New Zealand
| | - Mike Garratt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Ting Wang
- Department of Mathematics and Statistics, University of Otago, Dunedin, New Zealand
| | - John Hayes
- Department of Food Science, Penn State University, State College, PA, United States
| | - Mei Peng
- Sensory Neurosensory and Nutrtion Laboratory, Department of Food Science, University of Otago, Dunedin, New Zealand; Riddet Institute, Palmerston North, New Zealand.
| |
Collapse
|
31
|
He N, Liu M, Wu Y. Adipose tissue and hematopoiesis: Friend or foe? J Clin Lab Anal 2023; 37:e24872. [PMID: 36972475 PMCID: PMC10156104 DOI: 10.1002/jcla.24872] [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: 01/18/2023] [Revised: 03/02/2023] [Accepted: 03/12/2023] [Indexed: 03/29/2023] Open
Abstract
AIM Hematopoietic stem cells are the origin of all hematopoietic cells. They have the self-renewal ability and can differentiate into various blood cells. In physiological state, most of the hematopoietic stem cells are dormant, and only a few cells proliferate to maintain hematopoietic homeostasis. METHODS This precise steady-state maintenance is regulated by complex mechanisms. Bone marrow adipocytes make up half of all cells in the bone marrow cavity, a feature that has attracted the attention of researchers from multiple fields. The adipocyte density within marrow increases during aging and obesity. RESULTS Recent studies have shown that bone marrow adipocytes play important roles in regulating hematopoiesis, but the effects of bone marrow adipocytes on hematopoiesis are often conflicting. Bone marrow adipocytes, participating in the formation of bone marrow hematopoietic microenvironment, influence hematopoiesis positively or negatively. In addition, other adipose tissue, especially white adipose tissue, also regulates hematopoiesis. CONCLUSION In this review, we describe the role of adipose tissue in hematological malignancies, which may be useful for understanding hematopoiesis and the pathogenesis of related diseases.
Collapse
Affiliation(s)
- Na He
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Min Liu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yue Wu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
32
|
Stevens HE, Scuderi S, Collica SC, Tomasi S, Horvath TL, Vaccarino FM. Neonatal loss of FGFR2 in astroglial cells affects locomotion, sociability, working memory, and glia-neuron interactions in mice. Transl Psychiatry 2023; 13:89. [PMID: 36906620 PMCID: PMC10008554 DOI: 10.1038/s41398-023-02372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 03/13/2023] Open
Abstract
Fibroblast growth factor receptor 2 (FGFR2) is almost exclusively expressed in glial cells in postnatal mouse brain, but its impact in glia for brain behavioral functioning is poorly understood. We compared behavioral effects from FGFR2 loss in both neurons and astroglial cells and from FGFR2 loss in astroglial cells by using either the pluripotent progenitor-driven hGFAP-cre or the tamoxifen-inducible astrocyte-driven GFAP-creERT2 in Fgfr2 floxed mice. When FGFR2 was eliminated in embryonic pluripotent precursors or in early postnatal astroglia, mice were hyperactive, and had small changes in working memory, sociability, and anxiety-like behavior. In contrast, FGFR2 loss in astrocytes starting at 8 weeks of age resulted only in reduced anxiety-like behavior. Therefore, early postnatal loss of FGFR2 in astroglia is critical for broad behavioral dysregulation. Neurobiological assessments demonstrated that astrocyte-neuron membrane contact was reduced and glial glutamine synthetase expression increased only by early postnatal FGFR2 loss. We conclude that altered astroglial cell function dependent on FGFR2 in the early postnatal period may result in impaired synaptic development and behavioral regulation, modeling childhood behavioral deficits like attention deficit hyperactivity disorder (ADHD).
Collapse
Affiliation(s)
- Hanna E Stevens
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Psychiatry, Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, 52246, USA.
| | - Soraya Scuderi
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Sarah C Collica
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Simone Tomasi
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Tamas L Horvath
- Department of Neuroscience, Yale University, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Department of Obstetrics and Gynecology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Flora M Vaccarino
- Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Neuroscience, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
33
|
Fang X, Chen Y, Wang J, Zhang Z, Bai Y, Denney K, Gan L, Guo M, Weintraub NL, Lei Y, Lu XY. Increased intrinsic and synaptic excitability of hypothalamic POMC neurons underlies chronic stress-induced behavioral deficits. Mol Psychiatry 2023; 28:1365-1382. [PMID: 36473997 PMCID: PMC10005948 DOI: 10.1038/s41380-022-01872-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
Chronic stress exposure induces maladaptive behavioral responses and increases susceptibility to neuropsychiatric conditions. However, specific neuronal populations and circuits that are highly sensitive to stress and trigger maladaptive behavioral responses remain to be identified. Here we investigate the patterns of spontaneous activity of proopiomelanocortin (POMC) neurons in the arcuate nucleus (ARC) of the hypothalamus following exposure to chronic unpredictable stress (CUS) for 10 days, a stress paradigm used to induce behavioral deficits such as anhedonia and behavioral despair [1, 2]. CUS exposure increased spontaneous firing of POMC neurons in both male and female mice, attributable to reduced GABA-mediated synaptic inhibition and increased intrinsic neuronal excitability. While acute activation of POMC neurons failed to induce behavioral changes in non-stressed mice of both sexes, subacute (3 days) and chronic (10 days) repeated activation of POMC neurons was sufficient to induce anhedonia and behavioral despair in males but not females under non-stress conditions. Acute activation of POMC neurons promoted susceptibility to subthreshold unpredictable stress in both male and female mice. Conversely, acute inhibition of POMC neurons was sufficient to reverse CUS-induced anhedonia and behavioral despair in both sexes. Collectively, these results indicate that chronic stress induces both synaptic and intrinsic plasticity of POMC neurons, leading to neuronal hyperactivity. Our findings suggest that POMC neuron dysfunction drives chronic stress-related behavioral deficits.
Collapse
Affiliation(s)
- Xing Fang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Yuting Chen
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jiangong Wang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ziliang Zhang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Yu Bai
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Kirstyn Denney
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Lin Gan
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ming Guo
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Neal L Weintraub
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Yun Lei
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
34
|
Belaïdouni Y, Diabira D, Brosset-Heckel M, Valsamides V, Graziano JC, Santos C, Menuet C, Wayman GA, Gaiarsa JL. Leptin antagonism improves Rett syndrome phenotype in symptomatic male Mecp2-null mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.526251. [PMID: 36778454 PMCID: PMC9915649 DOI: 10.1101/2023.02.03.526251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder that arise from de novo mutations in the X-linked gene MECP2 (methyl-CpG-binding protein 2). Circulating levels of the adipocyte hormone leptin are elevated in RTT patients and rodent models of the disease. Leptin targets a large number of brain structures and regulates a wide range of developmental and physiological functions which are altered in RTT. We hypothesized that elevated leptin levels might contribute to RTT pathogenesis. Accordingly, we show that pharmacological antagonism of leptin or genetic reduction of leptin production prevents the degradation of health status, weight loss and the progression of breathing and locomotor deficits. At the neuronal level, the anti-leptin strategies rescue the hippocampal excitatory/inhibitory imbalance and synaptic plasticity impairment. Targeting leptin might therefore represent a new approach for RTT treatment.
Collapse
|
35
|
Liu H, He Y, Bai J, Zhang C, Zhang F, Yang Y, Luo H, Yu M, Liu H, Tu L, Zhang N, Yin N, Han J, Yan Z, Scarcelli NA, Conde KM, Wang M, Bean JC, Potts CHS, Wang C, Hu F, Liu F, Xu Y. Hypothalamic Grb10 enhances leptin signalling and promotes weight loss. Nat Metab 2023; 5:147-164. [PMID: 36593271 DOI: 10.1038/s42255-022-00701-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 10/19/2022] [Indexed: 01/04/2023]
Abstract
Leptin acts on hypothalamic neurons expressing agouti-related protein (AgRP) or pro-opiomelanocortin (POMC) to suppress appetite and increase energy expenditure, but the intracellular mechanisms that modulate central leptin signalling are not fully understood. Here we show that growth factor receptor-bound protein 10 (Grb10), an adaptor protein that binds to the insulin receptor and negatively regulates its signalling pathway, can interact with the leptin receptor and enhance leptin signalling. Ablation of Grb10 in AgRP neurons promotes weight gain, while overexpression of Grb10 in AgRP neurons reduces body weight in male and female mice. In parallel, deletion or overexpression of Grb10 in POMC neurons exacerbates or attenuates diet-induced obesity, respectively. Consistent with its role in leptin signalling, Grb10 in AgRP and POMC neurons enhances the anorexic and weight-reducing actions of leptin. Grb10 also exaggerates the inhibitory effects of leptin on AgRP neurons via ATP-sensitive potassium channel-mediated currents while facilitating the excitatory drive of leptin on POMC neurons through transient receptor potential channels. Our study identifies Grb10 as a potent leptin sensitizer that contributes to the maintenance of energy homeostasis by enhancing the response of AgRP and POMC neurons to leptin.
Collapse
Affiliation(s)
- Hailan Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Yang He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Juli Bai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Cell Systems & Anatomy and Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Chuanhai Zhang
- Department of Cell Systems & Anatomy and Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Feng Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Nan Zhang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Na Yin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Zili Yan
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Nikolas Anthony Scarcelli
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Kristine Marie Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Jonathan Carter Bean
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Camille Hollan Sidell Potts
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Fang Hu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
36
|
Qiu J, Bosch MA, Stincic TL, Hunker AC, Zweifel LS, Rønnekleiv OK, Kelly MJ. CRISPR/SaCas9 mutagenesis of stromal interaction molecule 1 in proopiomelanocortin neurons increases glutamatergic excitability and protects against diet-induced obesity. Mol Metab 2022; 66:101645. [PMID: 36442744 PMCID: PMC9727646 DOI: 10.1016/j.molmet.2022.101645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/09/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Proopiomelanocortin (POMC) neurons are the key anorexigenic hypothalamic neuron for integrating metabolic cues to generate the appropriate output for maintaining energy homeostasis and express the requisite channels as a perfect synaptic integrator in this role. Similar to the metabolic hormones leptin and insulin, glutamate also excites POMC neurons via group I metabotropic glutamate receptors (mGluR1 and 5, mGluR1/5) that activate Transient Receptor Potential Canonical (TRPC 5) Channels to cause depolarization. A key modulator of TRPC 5 channel activity is stromal interaction molecule 1 (STIM1), which is involved in recruitment of TRPC 5 channels from receptor-operated to store-operated calcium entry following depletion of calcium from the endoplasmic reticulum. METHODS We used a single adeno-associated viral (AAV) vector containing a recombinase-dependent Staphylococcus aureus Cas9 (SaCas) and a single guide RNA (sgRNA) to mutate Stim1 in POMCCre neurons in male mice, verified by qPCR of Stim1 mRNA expression in single POMC neurons. Whole-cell patch clamp experiments were conducted to validate the effects of Stim1 mutagenesis. Body weight and food intake were measured in male mice to assess disruptions in energy balance. RESULTS Reduced Stim1 expression augmented the efficacy of the mGluR1/5 agonist 3, 5-Dihydroxyphenylglycine (DHPG) to depolarize POMC neurons via a Gαq-coupled signaling pathway, which is an essential part of excitatory glutamatergic input in regulating energy homeostasis. The TRPC 5 channel blockers HC070 and Pico145 antagonized the excitatory effects of DHPG. As proof of principle, mutagenesis of Stim1 in POMC neurons reduced food intake, attenuated weight gain, reduced body fat and fat pad mass in mice fed a high fat diet. CONCLUSIONS Using CRISPR technology we have uncovered a critical role of STIM1 in modulating glutamatergic activation of TRPC 5 channels in POMC neurons, which ultimately is important for maintaining energy balance.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA,Corresponding author.Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Martha A. Bosch
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Todd L. Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Avery C. Hunker
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Larry S. Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA,Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Oline K. Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA,Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Martin J. Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA,Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA,Corresponding author.Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
37
|
Sayers S, Le N, Hernandez J, Mata‐Pacheco V, Wagner EJ. The vital role of arcuate nociceptin/orphanin FQ neurones in mounting an oestradiol-dependent adaptive response to negative energy balance via inhibition of nearby proopiomelanocortin neurones. J Physiol 2022; 600:4939-4961. [PMID: 36217719 PMCID: PMC9828807 DOI: 10.1113/jp283378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023] Open
Abstract
We tested the hypothesis that N/OFQ neurones in the arcuate nucleus (N/OFQARC ) inhibit proopiomelanocortin (POMCARC ) neurones in a diet- and hormone-dependent manner to promote a more extensive rebound hyperphagia upon re-feeding following an 18 h fast. We utilized intact male or ovariectomized (OVX) female mice subjected to ad libitum-feeding or fasting conditions. N/OFQARC neurones under negative energy balance conditions displayed heightened sensitivity as evidenced by a decreased rheobase threshold, increased firing frequency, and increased burst duration and frequency compared to ad libitum-feeding conditions. Stimulation of N/OFQARC neurones more robustly inhibited POMCARC neurones under fasting conditions compared to ad libitum-feeding conditions. N/OFQARC inhibition of POMCARC neurones is hormone dependent as chemostimulation of N/OFQARC neurones from fasted males and OVX females produced a sizable outward current in POMCARC neurones. Oestradiol (E2 ) markedly attenuated the N/OFQ-induced POMCARC outward current. Additionally, N/OFQ tonically inhibits POMCARC neurones to a greater degree under fasting conditions than in ad libitum-feeding conditions as evidenced by the abrogation of N/OFQ-nociceptin opioid peptide (NOP) receptor signalling and inhibition of N/OFQ release via chemoinhibition of N/OFQARC neurones. Intra-arcuate nucleus application of N/OFQ further elevated the hyperphagic response and increased meal size during the 6 h re-feed period, and these effects were mimicked by chemostimulation of N/OFQARC neurones in vivo. E2 attenuated the robust N/OFQ-induced rebound hyperphagia seen in vehicle-treated OVX females. These data demonstrate that N/OFQARC neurones play a vital role in mitigating the impact of negative energy balance by inhibiting the excitability of anorexigenic neural substrates, an effect that is diminished by E2 in females. KEY POINTS: Nociceptin/orphanin FQ (N/OFQ) promotes increased energy intake and decreased energy expenditure under conditions of positive energy balance in a sex- and hormone-dependent manner. Here it is shown that under conditions of negative energy balance, i.e. fasting, N/OFQ inhibits anorexigenic proopiomelanocortin (POMC) neurones to a greater degree compared to homeostatic conditions due to fasting-induced hyperexcitability of N/OFQ neurones. Additionally, N/OFQ promotes a sustained increase in rebound hyperphagia and increase in meal size during the re-feed period following a fast. These results promote greater understanding of how energy balance influences the anorexigenic circuitry of the hypothalamus, and aid in understanding the neurophysiological pathways implicated in eating disorders promoting cachexia.
Collapse
Affiliation(s)
- Sarah Sayers
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Nikki Le
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Jennifer Hernandez
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Veronica Mata‐Pacheco
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Edward J. Wagner
- College of Osteopathic Medicine of the PacificWestern University of Health SciencesPomonaCAUSA
| |
Collapse
|
38
|
Xie D, Stutz B, Li F, Chen F, Lv H, Sestan-Pesa M, Catarino J, Gu J, Zhao H, Stoddard CE, Carmichael GG, Shanabrough M, Taylor HS, Liu ZW, Gao XB, Horvath TL, Huang Y. TET3 epigenetically controls feeding and stress response behaviors via AGRP neurons. J Clin Invest 2022; 132:162365. [PMID: 36189793 PMCID: PMC9525119 DOI: 10.1172/jci162365] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
The TET family of dioxygenases promote DNA demethylation by oxidizing 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). Hypothalamic agouti-related peptide-expressing (AGRP-expressing) neurons play an essential role in driving feeding, while also modulating nonfeeding behaviors. Besides AGRP, these neurons produce neuropeptide Y (NPY) and the neurotransmitter GABA, which act in concert to stimulate food intake and decrease energy expenditure. Notably, AGRP, NPY, and GABA can also elicit anxiolytic effects. Here, we report that in adult mouse AGRP neurons, CRISPR-mediated genetic ablation of Tet3, not previously known to be involved in central control of appetite and metabolism, induced hyperphagia, obesity, and diabetes, in addition to a reduction of stress-like behaviors. TET3 deficiency activated AGRP neurons, simultaneously upregulated the expression of Agrp, Npy, and the vesicular GABA transporter Slc32a1, and impeded leptin signaling. In particular, we uncovered a dynamic association of TET3 with the Agrp promoter in response to leptin signaling, which induced 5hmC modification that was associated with a chromatin-modifying complex leading to transcription inhibition, and this regulation occurred in both the mouse models and human cells. Our results unmasked TET3 as a critical central regulator of appetite and energy metabolism and revealed its unexpected dual role in the control of feeding and other complex behaviors through AGRP neurons.
Collapse
Affiliation(s)
- Di Xie
- Department of Obstetrics, Gynecology and Reproductive Sciences.,Yale Center for Molecular and Systems Metabolism, and
| | - Bernardo Stutz
- Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Feng Li
- Department of Obstetrics, Gynecology and Reproductive Sciences.,Yale Center for Molecular and Systems Metabolism, and
| | - Fan Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences
| | - Haining Lv
- Department of Obstetrics, Gynecology and Reproductive Sciences.,Yale Center for Molecular and Systems Metabolism, and
| | - Matija Sestan-Pesa
- Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jonatas Catarino
- Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jianlei Gu
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Christopher E Stoddard
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Gordon G Carmichael
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marya Shanabrough
- Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences
| | - Zhong-Wu Liu
- Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xiao-Bing Gao
- Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tamas L Horvath
- Department of Obstetrics, Gynecology and Reproductive Sciences.,Yale Center for Molecular and Systems Metabolism, and.,Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yingqun Huang
- Department of Obstetrics, Gynecology and Reproductive Sciences.,Yale Center for Molecular and Systems Metabolism, and
| |
Collapse
|
39
|
Boutagouga Boudjadja M, Culotta I, De Paula GC, Harno E, Hunter J, Cavalcanti-de-Albuquerque JP, Luckman SM, Hepworth M, White A, Aviello G, D'Agostino G. Hypothalamic AgRP neurons exert top-down control on systemic TNF-α release during endotoxemia. Curr Biol 2022; 32:4699-4706.e4. [PMID: 36182699 DOI: 10.1016/j.cub.2022.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/21/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022]
Abstract
Loss of appetite and negative energy balance are common features of endotoxemia in all animals and are thought to have protective roles by reducing nutrient availability to host and pathogen metabolism. Accordingly, fasting and caloric restriction have well-established anti-inflammatory properties. However, in response to reduced nutrient availability at the cellular and organ levels, negative energy balance also recruits distinct energy-sensing brain circuits, but it is not known whether these neuronal systems have a role in its anti-inflammatory effects. Here, we report that hypothalamic AgRP neurons-a critical neuronal population for the central representation of negative energy balance-have parallel immunoregulatory functions. We found that when endotoxemia occurs in fasted mice, the activity of AgRP neurons remains sustained, but this activity does not influence feeding behavior and endotoxemic anorexia. Furthermore, we found that endotoxemia acutely desensitizes AgRP neurons, which also become refractory to inhibitory signals. Mimicking this sustained AgRP neuron activity in fed mice by chemogenetic activation-a manipulation known to recapitulate core behavioral features of fasting-results in reduced acute tumor necrosis factor alpha (TNF-α) release during endotoxemia. Mechanistically, we found that endogenous glucocorticoids play an important role: glucocorticoid receptor deletion from AgRP neurons prevents their endotoxemia-induced desensitization, and importantly, it counteracts the fasting-induced suppression of TNF-α release, resulting in prolonged sickness. Together, these findings provide evidence directly linking AgRP neuron activity to the acute response during endotoxemia, suggesting that these neurons are a functional component of the immunoregulatory effects associated with negative energy balance and catabolic metabolism.
Collapse
Affiliation(s)
- Mehdi Boutagouga Boudjadja
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Isabella Culotta
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Erika Harno
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jenna Hunter
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Simon M Luckman
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Matthew Hepworth
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Anne White
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Gabriella Aviello
- The Rowett Institute, University of Aberdeen, Aberdeen, UK; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Giuseppe D'Agostino
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; The Rowett Institute, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
40
|
Hwang E, Scarlett JM, Baquero AF, Bennett CM, Dong Y, Chau D, Brown JM, Mercer AJ, Meek TH, Grove KL, Phan BAN, Morton GJ, Williams KW, Schwartz MW. Sustained inhibition of NPY/AgRP neuronal activity by FGF1. JCI Insight 2022; 7:e160891. [PMID: 35917179 PMCID: PMC9536267 DOI: 10.1172/jci.insight.160891] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
In rodent models of type 2 diabetes (T2D), central administration of FGF1 normalizes elevated blood glucose levels in a manner that is sustained for weeks or months. Increased activity of NPY/AgRP neurons in the hypothalamic arcuate nucleus (ARC) is implicated in the pathogenesis of hyperglycemia in these animals, and the ARC is a key brain area for the antidiabetic action of FGF1. We therefore sought to determine whether FGF1 inhibits NPY/AgRP neurons and, if so, whether this inhibitory effect is sufficiently durable to offer a feasible explanation for sustained diabetes remission induced by central administration of FGF1. Here, we show that FGF1 inhibited ARC NPY/AgRP neuron activity, both after intracerebroventricular injection in vivo and when applied ex vivo in a slice preparation; we also showed that the underlying mechanism involved increased input from presynaptic GABAergic neurons. Following central administration, the inhibitory effect of FGF1 on NPY/AgRP neurons was also highly durable, lasting for at least 2 weeks. To our knowledge, no precedent for such a prolonged inhibitory effect exists. Future studies are warranted to determine whether NPY/AgRP neuron inhibition contributes to the sustained antidiabetic action elicited by intracerebroventricular FGF1 injection in rodent models of T2D.
Collapse
Affiliation(s)
- Eunsang Hwang
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Jarrad M. Scarlett
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, Washington, USA
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Arian F. Baquero
- Obesity Research, Novo Nordisk Research Center Seattle, Seattle, Washington, USA
| | - Camdin M. Bennett
- Obesity Research, Novo Nordisk Research Center Seattle, Seattle, Washington, USA
| | - Yanbin Dong
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Dominic Chau
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Jenny M. Brown
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, Washington, USA
- University of Copenhagen, Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Aaron J. Mercer
- Obesity Research, Novo Nordisk Research Center Seattle, Seattle, Washington, USA
| | - Thomas H. Meek
- Obesity Research, Novo Nordisk Research Center Seattle, Seattle, Washington, USA
- Discovery Technologies & Genomics, Novo Nordisk Research Centre Oxford, Oxford, United Kingdom
| | - Kevin L. Grove
- Obesity Research, Novo Nordisk Research Center Seattle, Seattle, Washington, USA
| | - Bao Anh N. Phan
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, Washington, USA
| | - Gregory J. Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, Washington, USA
| | - Kevin W. Williams
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Michael W. Schwartz
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, Washington, USA
| |
Collapse
|
41
|
Hebebrand J, Hildebrandt T, Schlögl H, Seitz J, Denecke S, Vieira D, Gradl-Dietsch G, Peters T, Antel J, Lau D, Fulton S. The role of hypoleptinemia in the psychological and behavioral adaptation to starvation: implications for anorexia nervosa. Neurosci Biobehav Rev 2022; 141:104807. [PMID: 35931221 DOI: 10.1016/j.neubiorev.2022.104807] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/11/2022] [Accepted: 07/31/2022] [Indexed: 12/17/2022]
Abstract
This narrative review aims to pinpoint mental and behavioral effects of starvation, which may be triggered by hypoleptinemia and as such may be amenable to treatment with leptin receptor agonists. The reduced leptin secretion results from the continuous loss of fat mass, thus initiating a graded triggering of diverse starvation related adaptive functions. In light of leptin receptors located in several peripheral tissues and many brain regions adaptations may extend beyond those of the hypothalamus-pituitary-end organ-axes. We focus on gastrointestinal tract and reward system as relevant examples of peripheral and central effects of leptin. Despite its association with extreme obesity, congenital leptin deficiency with its many parallels to a state of starvation allows the elucidation of mental symptoms amenable to treatment with exogenous leptin in both ob/ob mice and humans with this autosomal recessive disorder. For starvation induced behavioral changes with an intact leptin signaling we particularly focus on rodent models for which proof of concept has been provided for the causative role of hypoleptinemia. For humans, we highlight the major cognitive, emotional and behavioral findings of the Minnesota Starvation Experiment to contrast them with results obtained upon a lesser degree of caloric restriction. Evidence for hypoleptinemia induced mental changes also stems from findings obtained in lipodystrophies. In light of the recently reported beneficial cognitive, emotional and behavioral effects of metreleptin-administration in anorexia nervosa we discuss potential implications for the treatment of this eating disorder. We postulate that leptin has profound psychopharmacological effects in the state of starvation.
Collapse
Affiliation(s)
- Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Tom Hildebrandt
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Haiko Schlögl
- Department of Endocrinology, Nephrology, Rheumatology, Division of Endocrinology, University Hospital Leipzig, Liebigstr. 20, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH University Hospital Aachen, Germany
| | - Saskia Denecke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Diana Vieira
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Gertraud Gradl-Dietsch
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Triinu Peters
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - David Lau
- Department of Nutrition, Neuroscience - University of Montreal & CRCHUM, Montréal QC H3T1J4, Canada
| | - Stephanie Fulton
- Department of Nutrition, Neuroscience - University of Montreal & CRCHUM, Montréal QC H3T1J4, Canada
| |
Collapse
|
42
|
Benevento M, Hökfelt T, Harkany T. Ontogenetic rules for the molecular diversification of hypothalamic neurons. Nat Rev Neurosci 2022; 23:611-627. [PMID: 35906427 DOI: 10.1038/s41583-022-00615-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
The hypothalamus is an evolutionarily conserved endocrine interface that, among other roles, links central homeostatic control to adaptive bodily responses by releasing hormones and neuropeptides from its many neuronal subtypes. In its preoptic, anterior, tuberal and mammillary subdivisions, a kaleidoscope of magnocellular and parvocellular neuroendocrine command neurons, local-circuit neurons, and neurons that project to extrahypothalamic areas are intermingled in partially overlapping patches of nuclei. Molecular fingerprinting has produced data of unprecedented mass and depth to distinguish and even to predict the synaptic and endocrine competences, connectivity and stimulus selectivity of many neuronal modalities. These new insights support eminent studies from the past century but challenge others on the molecular rules that shape the developmental segregation of hypothalamic neuronal subtypes and their use of morphogenic cues for terminal differentiation. Here, we integrate single-cell RNA sequencing studies with those of mouse genetics and endocrinology to describe key stages of hypothalamus development, including local neurogenesis, the direct terminal differentiation of glutamatergic neurons, transition cascades for GABAergic and GABAergic cell-derived dopamine cells, waves of local neuronal migration, and sequential enrichment in neuropeptides and hormones. We particularly emphasize how transcription factors determine neuronal identity and, consequently, circuit architecture, and whether their deviations triggered by environmental factors and hormones provoke neuroendocrine illnesses.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria. .,Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
43
|
Duncan PJ, McClafferty H, Nolan O, Ding Q, Homer NZM, Le Tissier P, Walker BR, Shipston MJ, Romanò N, Chambers TJG. Corticotroph isolation from Pomc-eGFP mice reveals sustained transcriptional dysregulation characterising a mouse model of glucocorticoid-induced suppression of the hypothalamus-pituitary-adrenal axis. J Neuroendocrinol 2022; 34:e13165. [PMID: 35833423 PMCID: PMC9539609 DOI: 10.1111/jne.13165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 11/27/2022]
Abstract
Glucocorticoids (GC) are prescribed for periods > 3 months to 1%-3% of the UK population; 10%-50% of these patients develop hypothalamus-pituitary-adrenal (HPA) axis suppression, which may last over 6 months and is associated with morbidity and mortality. Recovery of the pituitary and hypothalamus is necessary for recovery of adrenal function. We developed a mouse model of dexamethasone (DEX)-induced HPA axis dysfunction aiming to further explore recovery in the pituitary. Adult male wild-type C57BL6/J or Pomc-eGFP transgenic mice were randomly assigned to receive DEX (approximately 0.4 mg kg-1 bodyweight day-1 ) or vehicle via drinking water for 4 weeks following which treatment was withdrawn and tissues were harvested after another 0, 1, and 4 weeks. Corticotrophs were isolated from Pomc-eGFP pituitaries using fluorescence-activated cell sorting, and RNA extracted for RNA-sequencing. DEX treatment suppressed corticosterone production, which remained partially suppressed at least 1 week following DEX withdrawal. In the adrenal, Hsd3b2, Cyp11a1, and Mc2r mRNA levels were significantly reduced at time 0, with Mc2r and Cyp11a1 remaining reduced 1 week following DEX withdrawal. The corticotroph transcriptome was modified by DEX treatment, with some differences between groups persisting 4 weeks following withdrawal. No genes supressed by DEX exhibited ongoing attenuation 1 and 4 weeks following withdrawal, whereas only two genes were upregulated and remained so following withdrawal. A pattern of rebound at 1 and 4 weeks was observed in 14 genes that increased following suppression, and in six genes that were reduced by DEX and then increased. Chronic GC treatment may induce persistent changes in the pituitary that may influence future response to GC treatment or stress.
Collapse
Affiliation(s)
- Peter J. Duncan
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | | | - Oscar Nolan
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Qinghui Ding
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Natalie Z. M. Homer
- Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research InstituteEdinburghUK
| | - Paul Le Tissier
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Brian R. Walker
- Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research InstituteEdinburghUK
- Translational & Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | - Nicola Romanò
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Thomas J. G. Chambers
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- Edinburgh Centre for Endocrinology and DiabetesNHS Lothian, Metabolic Unit, Western General HospitalEdinburghUK
| |
Collapse
|
44
|
Ryu Y, Kim Y, Lim HR, Kim HJ, Park BS, Kim JG, Park SJ, Ha CM. Single-Step Fast Tissue Clearing of Thick Mouse Brain Tissue for Multi-Dimensional High-Resolution Imaging. Int J Mol Sci 2022; 23:ijms23126826. [PMID: 35743267 PMCID: PMC9224586 DOI: 10.3390/ijms23126826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/10/2022] Open
Abstract
Recent advances in optical clearing techniques have dramatically improved deep tissue imaging by reducing the obscuring effects of light scattering and absorption. However, these optical clearing methods require specialized equipment or a lengthy undertaking with complex protocols that can lead to sample volume changes and distortion. In addition, the imaging of cleared tissues has limitations, such as fluorescence bleaching, harmful and foul-smelling solutions, and the difficulty of handling samples in high-viscosity refractive index (RI) matching solutions. To address the various limitations of thick tissue imaging, we developed an Aqueous high refractive Index matching and tissue Clearing solution for Imaging (termed AICI) with a one-step tissue clearing protocol that was easily made at a reasonable price in our own laboratory without any equipment. AICI can rapidly clear a 1 mm thick brain slice within 90 min with simultaneous RI matching, low viscosity, and a high refractive index (RI = 1.466), allowing the imaging of the sample without additional processing. We compared AICI with commercially available RI matching solutions, including optical clear agents (OCAs), for tissue clearing. The viscosity of AICI is closer to that of water compared with other RI matching solutions, and there was a less than 2.3% expansion in the tissue linear morphology during 24 h exposure to AICI. Moreover, AICI remained fluid over 30 days of air exposure, and the EGFP fluorescence signal was only reduced to ~65% after 10 days. AICI showed a limited clearing of brain tissue >3 mm thick. However, fine neuronal structures, such as dendritic spines and axonal boutons, could still be imaged in thick brain slices treated with AICI. Therefore, AICI is useful not only for the three-dimensional (3D) high-resolution identification of neuronal structures, but also for the examination of multiple structural imaging by neuronal distribution, projection, and gene expression in deep brain tissue. AICI is applicable beyond the imaging of fluorescent antibodies and dyes, and can clear a variety of tissue types, making it broadly useful to researchers for optical imaging applications.
Collapse
Affiliation(s)
- Youngjae Ryu
- Research Strategy Office and Brain Research Core Facilities of Korea Brain Research Institute, Daegu 41068, Korea; (Y.R.); (Y.K.); (H.R.L.)
- Department of Histology, College of Veterinary Medicine, Kyungpook University, Daegu 41566, Korea;
| | - Yoonju Kim
- Research Strategy Office and Brain Research Core Facilities of Korea Brain Research Institute, Daegu 41068, Korea; (Y.R.); (Y.K.); (H.R.L.)
| | - Hye Ryeong Lim
- Research Strategy Office and Brain Research Core Facilities of Korea Brain Research Institute, Daegu 41068, Korea; (Y.R.); (Y.K.); (H.R.L.)
| | - Hyung-Joon Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu 41068, Korea;
| | - Byong Seo Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (B.S.P.); (J.G.K.)
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (B.S.P.); (J.G.K.)
| | - Sang-Joon Park
- Department of Histology, College of Veterinary Medicine, Kyungpook University, Daegu 41566, Korea;
| | - Chang Man Ha
- Research Strategy Office and Brain Research Core Facilities of Korea Brain Research Institute, Daegu 41068, Korea; (Y.R.); (Y.K.); (H.R.L.)
- Correspondence:
| |
Collapse
|
45
|
Harvey J. Food for Thought: Leptin and Hippocampal Synaptic Function. Front Pharmacol 2022; 13:882158. [PMID: 35784728 PMCID: PMC9247348 DOI: 10.3389/fphar.2022.882158] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
It is well documented that the endocrine hormone, leptin controls energy homeostasis by providing key signals to specific hypothalamic nuclei. However, our knowledge of leptin’s central actions has advanced considerably over the last 20 years, with the hippocampus now established as an important brain target for this hormone. Leptin receptors are highly localised to hippocampal synapses, and increasing evidence reveals that activation of synaptically located leptin receptors markedly impacts cognitive processes, and specifically hippocampal-dependent learning and memory. Here, we review the recent actions of leptin at hippocampal synapses and explore the consequences for brain health and disease.
Collapse
|
46
|
Banerjee J, Dorfman MD, Fasnacht R, Douglass JD, Wyse-Jackson AC, Barria A, Thaler JP. CX3CL1 Action on Microglia Protects from Diet-Induced Obesity by Restoring POMC Neuronal Excitability and Melanocortin System Activity Impaired by High-Fat Diet Feeding. Int J Mol Sci 2022; 23:6380. [PMID: 35742824 PMCID: PMC9224384 DOI: 10.3390/ijms23126380] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/31/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
Both hypothalamic microglial inflammation and melanocortin pathway dysfunction contribute to diet-induced obesity (DIO) pathogenesis. Previous studies involving models of altered microglial signaling demonstrate altered DIO susceptibility with corresponding POMC neuron cytological changes, suggesting a link between microglia and the melanocortin system. We addressed this hypothesis using the specific microglial silencing molecule, CX3CL1 (fractalkine), to determine whether reducing hypothalamic microglial activation can restore POMC/melanocortin signaling to protect against DIO. We performed metabolic analyses in high fat diet (HFD)-fed mice with targeted viral overexpression of CX3CL1 in the hypothalamus. Electrophysiologic recording in hypothalamic slices from POMC-MAPT-GFP mice was used to determine the effects of HFD feeding and microglial silencing via minocycline or CX3CL1 on GFP-labeled POMC neurons. Finally, mice with hypothalamic overexpression of CX3CL1 received central treatment with the melanocortin receptor antagonist SHU9119 to determine whether melanocortin signaling is required for the metabolic benefits of CX3CL1. Hypothalamic overexpression of CX3CL1 increased leptin sensitivity and POMC gene expression, while reducing weight gain in animals fed an HFD. In electrophysiological recordings from hypothalamic slice preparations, HFD feeding was associated with reduced POMC neuron excitability and increased amplitude of inhibitory postsynaptic currents. Microglial silencing using minocycline or CX3CL1 treatment reversed these HFD-induced changes in POMC neuron electrophysiologic properties. Correspondingly, blockade of melanocortin receptor signaling in vivo prevented both the acute and chronic reduction in food intake and body weight mediated by CX3CL1. Our results show that suppressing microglial activation during HFD feeding reduces DIO susceptibility via a mechanism involving increased POMC neuron excitability and melanocortin signaling.
Collapse
Affiliation(s)
- Jineta Banerjee
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; (J.B.); (M.D.D.); (R.F.); (J.D.D.); (A.C.W.-J.)
| | - Mauricio D. Dorfman
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; (J.B.); (M.D.D.); (R.F.); (J.D.D.); (A.C.W.-J.)
| | - Rachael Fasnacht
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; (J.B.); (M.D.D.); (R.F.); (J.D.D.); (A.C.W.-J.)
| | - John D. Douglass
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; (J.B.); (M.D.D.); (R.F.); (J.D.D.); (A.C.W.-J.)
| | - Alice C. Wyse-Jackson
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; (J.B.); (M.D.D.); (R.F.); (J.D.D.); (A.C.W.-J.)
| | - Andres Barria
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98109, USA;
| | - Joshua P. Thaler
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; (J.B.); (M.D.D.); (R.F.); (J.D.D.); (A.C.W.-J.)
| |
Collapse
|
47
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
48
|
Lee W, Dwortz MF, Milewski TM, Champagne FA, Curley JP. Social status mediated variation in hypothalamic transcriptional profiles of male mice. Horm Behav 2022; 142:105176. [PMID: 35500322 DOI: 10.1016/j.yhbeh.2022.105176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/07/2022] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
Animals of different social status exhibit variation in aggression, territorial and reproductive behavior as well as activity patterns, feeding, drinking and status signaling. This behavioral and physiological plasticity is coordinated by underlying changes in brain gene transcription. Using Tag-based RNA sequencing (Tag-seq), we explore RNA transcriptomes from the medial preoptic area (mPOA) and ventral hypothalamus (vHYP) of male mice of different social ranks in a dominance hierarchy and detect candidate genes and cellular pathways that underlie status-related plasticity. Within the mPOA, oxytocin (Oxt) and vasopressin (Avp) are more highly expressed in subdominant mice compared to other ranks, while nitric oxide synthase (Nos1) has lower expression in subdominant mice. Within the vHYP, we find that both orexigenic and anorexigenic genes involved in feeding behavior, including agouti-related peptide (Agrp), neuropeptide-Y (Npy), galanin (Gal), proopiomelanocortin (Pomc), and Cocaine- and Amphetamine-Regulated Transcript Protein prepropeptide (Cartpt), are less expressed in dominant animals compared to more subordinate ranks. We suggest that this may represent a reshaping of feeding circuits in dominant compared to subdominant and subordinate animals. Furthermore, we determine several genes that are positively and negatively associated with the level of despotism (aggression) in dominant males. Ultimately, we identify hypothalamic genes controlling feeding and social behaviors that are differentially transcribed across animals of varying social status. These changes in brain transcriptomics likely support phenotypic variation that enable animals to adapt to their current social status.
Collapse
Affiliation(s)
- W Lee
- Department of Psychology, University of Texas at Austin, Austin, TX, USA; Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, CA, USA
| | - M F Dwortz
- Department of Psychology, University of Texas at Austin, Austin, TX, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - T M Milewski
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - F A Champagne
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - J P Curley
- Department of Psychology, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
49
|
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: 1.0] [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
|
50
|
Development of "Hunger Neurons" and the Unanticipated Relationship Between Energy Metabolism and Mother-Infant Interactions. Biol Psychiatry 2022; 91:907-914. [PMID: 35397878 PMCID: PMC10184517 DOI: 10.1016/j.biopsych.2022.02.962] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 12/22/2022]
Abstract
Over the course of a lifetime, the perinatal period plays an outsized role in the function of physiological systems. Here, we discuss how neurons that regulate energy metabolism contribute to the infant's relationship with the mother. We focus our discussion on Agrp neurons, which are located in the arcuate nucleus of the hypothalamus. These neurons heavily regulate energy metabolism. Because offspring transition from a period of dependence on the caregiver to independence, we discuss the importance of the caregiver-offspring relationship for the function of Agrp neurons. We present evidence that in the adult, Agrp neurons motivate the animal to eat, while in the neonate, they motivate the offspring to seek the proximity of the caregiver. We specifically highlight the peculiarities in the development of Agrp neurons and how they relate to the regulation of metabolism and behavior over the course of a lifetime. In sum, this review considers the unique insights that ontogenetic studies can offer toward our understanding of complex biological systems, such as the regulation of energy metabolism and mother-infant attachment.
Collapse
|