1
|
Mehrabi H, Bansal P, Jutoy J, Chan YH, Roitman MF, Gao R, Jung EE. Optogenetic activation of hypothalamic AgRP neurons in transgenic zebrafish larvae increased food intake. Sci Rep 2025; 15:17976. [PMID: 40410389 PMCID: PMC12102260 DOI: 10.1038/s41598-025-03040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
Agouti Related Peptide (AgRP) neurons are located in the hypothalamus, and upon stimulation, these neurons regulate hunger and hunger-mediated behaviors, especially food-seeking and compulsive eating. AgRP neurons are naturally activated by ghrelin binding onto the ghrelin receptors on the neuron surface during starvation or fasting state to evoke the aforementioned behaviors. In this study, we used channelrhodopsin (ChR2), an optogenetic actuator, to control AgRP neuronal activity. For the first time, we observed food-intake behavior in zebrafish larvae by optogenetically triggering AgRP1 neurons. We created a transgenic line, Tg(AgRP1:ChR2-Kaede), where ChR2-Kaede is expressed in AgRP1 neurons. Transgenic zebrafish Tg(AgRP1:ChR2-Kaede) larvae at 6 days post fertilization and wild-type (ABWT) larvae were used to compare the suction behavior. We found that AgRP1 neuron activation in transgenic larvae led to a significantly higher food-consumption behavior than wildtype larvae when analyzed using Particle Image Velocimetry (PIV) to calculate the food particle velocity initiated by larval suction behavior. These findings in this novel transgenic zebrafish model would be useful in studying various hunger-related behaviors, their underlying neural circuits, and substrates subjected to different chemical stimuli, including drugs of abuse.
Collapse
Affiliation(s)
- Hossein Mehrabi
- Department of Mechanical Engineering, University of Illinois Chicago, 842 W Taylor St, Chicago, IL, 60607, USA
| | - Pushkar Bansal
- Department of Pharmacology and Toxicology, The University of Utah, 30S 2000E, Salt Lake City, UT, 84112, USA
| | - John Jutoy
- Department of Mechanical Engineering, University of Illinois Chicago, 842 W Taylor St, Chicago, IL, 60607, USA
| | - Yat Ho Chan
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor St, Chicago, IL, 60607, USA
| | - Mitchell F Roitman
- Department of Psychology, University of Illinois Chicago, 1007 W Harrison St, Chicago, IL, 60607, USA
| | - Ruixuan Gao
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor St, Chicago, IL, 60607, USA
- Department of Biological Sciences, University of Illinois Chicago, 845 W Taylor St, Chicago, IL, 60607, USA
| | - Erica E Jung
- Department of Mechanical Engineering, University of Illinois Chicago, 842 W Taylor St, Chicago, IL, 60607, USA.
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, 851 S Morgan St, Chicago, IL, 60607, USA.
| |
Collapse
|
2
|
Solheim MH, Stroganov S, Chen W, Subagia PS, Bauder CA, Wnuk-Lipinski D, Del Río-Martín A, Sotelo-Hitschfeld T, Beddows CA, Klemm P, Dodd GT, Lundh S, Secher A, Wunderlich FT, Steuernagel L, Brüning JC. Hypothalamic PNOC/NPY neurons constitute mediators of leptin-controlled energy homeostasis. Cell 2025:S0092-8674(25)00403-9. [PMID: 40273910 DOI: 10.1016/j.cell.2025.04.001] [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: 03/27/2024] [Revised: 12/23/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Leptin acts in the brain to suppress appetite, yet the responsible neurocircuitries underlying leptin's anorectic effect are incompletely defined. Prepronociceptin (PNOC)-expressing neurons mediate diet-induced hyperphagia and weight gain in mice. Here, we show that leptin regulates appetite and body weight via PNOC neurons, and that loss of leptin receptor (Lepr) expression in PNOC-expressing neurons in the arcuate nucleus of the hypothalamus (ARC) causes hyperphagia and obesity. Restoring Lepr expression in PNOC neurons on a Lepr-null obese background substantially reduces body weight. Lepr inactivation in PNOC neurons increases neuropeptide Y (Npy) expression in a subset of hypothalamic PNOC neurons that do not express agouti-related peptide (Agrp). Selective chemogenetic activation of PNOC/NPY neurons promotes feeding to the same extent as activating all PNOCARC neurons, and overexpression of Npy in PNOCARC neurons promotes hyperphagia and obesity. Thus, we introduce PNOC/NPYARC neurons as an additional critical mediator of leptin action and as a promising target for obesity therapeutics.
Collapse
Affiliation(s)
- Marie H Solheim
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Sima Stroganov
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - P Sicilia Subagia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Daria Wnuk-Lipinski
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Tamara Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Cait A Beddows
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, VIC, Australia
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Garron T Dodd
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, VIC, Australia
| | - Sofia Lundh
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Anna Secher
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Neurogenomics Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
3
|
Chen J, Cai M, Zhan C. Neuronal Regulation of Feeding and Energy Metabolism: A Focus on the Hypothalamus and Brainstem. Neurosci Bull 2025; 41:665-675. [PMID: 39704987 PMCID: PMC11978587 DOI: 10.1007/s12264-024-01335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/19/2024] [Indexed: 12/21/2024] Open
Abstract
In the face of constantly changing environments, the central nervous system (CNS) rapidly and accurately calculates the body's needs, regulates feeding behavior, and maintains energy homeostasis. The arcuate nucleus of the hypothalamus (ARC) plays a key role in this process, serving as a critical brain region for detecting nutrition-related hormones and regulating appetite and energy homeostasis. Agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons in the ARC are core elements that interact with other brain regions through a complex appetite-regulating network to comprehensively control energy homeostasis. In this review, we explore the discovery and research progress of AgRP neurons in regulating feeding and energy metabolism. In addition, recent advances in terms of feeding behavior and energy homeostasis, along with the redundant neural mechanisms involved in energy metabolism, are discussed. Finally, the challenges and opportunities in the field of neural regulation of feeding and energy metabolism are briefly discussed.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Meiting Cai
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Cheng Zhan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
4
|
Sun X, Liu B, Yuan Y, Rong Y, Pang R, Li Q. Neural and hormonal mechanisms of appetite regulation during eating. Front Nutr 2025; 12:1484827. [PMID: 40201582 PMCID: PMC11977392 DOI: 10.3389/fnut.2025.1484827] [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: 08/22/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Numerous animal and clinical studies have demonstrated that the arcuate nucleus of the hypothalamus, a central regulator of appetite, plays a significant role in modulating feeding behavior. However, current research primarily focuses on long-term dietary changes and their effects on the body, with limited investigation into neuroendocrine dynamics during individual meals across diverse populations. In contrast to long-term dietary adjustments, directives for dietary behavior during a specific meal are more actionable, potentially enhancing patient adherence and achieving better outcomes in dietary behavior interventions. This review aimed to explore the neural pathways and endocrine changes activated by gastrointestinal expansion and variations in blood nutrient levels during a single meal, with the goal of informing dietary behavior guidance.
Collapse
Affiliation(s)
- Xurui Sun
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Binghan Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuan Yuan
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Rong
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rui Pang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiu Li
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
5
|
Liang TZ, Jin ZY, Lin YJ, Chen ZY, Li Y, Xu JK, Yang F, Qin L. Targeting the central and peripheral nervous system to regulate bone homeostasis: mechanisms and potential therapies. Mil Med Res 2025; 12:13. [PMID: 40108680 PMCID: PMC11924829 DOI: 10.1186/s40779-025-00600-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
The skeleton is innervated by different types of nerves and receives signaling from the nervous system to maintain homeostasis and facilitate regeneration or repair. Although the role of peripheral nerves and signals in regulating bone homeostasis has been extensively investigated, the intimate relationship between the central nervous system and bone remains less understood, yet it has emerged as a hot topic in the bone field. In this review, we discussed clinical observations and animal studies that elucidate the connection between the nervous system and bone metabolism, either intact or after injury. First, we explored mechanistic studies linking specific brain nuclei with bone homeostasis, including the ventromedial hypothalamus, arcuate nucleus, paraventricular hypothalamic nucleus, amygdala, and locus coeruleus. We then focused on the characteristics of bone innervation and nerve subtypes, such as sensory, sympathetic, and parasympathetic nerves. Moreover, we summarized the molecular features and regulatory functions of these nerves. Finally, we included available translational approaches that utilize nerve function to improve bone homeostasis and promote bone regeneration. Therefore, considering the nervous system within the context of neuromusculoskeletal interactions can deepen our understanding of skeletal homeostasis and repair process, ultimately benefiting future clinical translation.
Collapse
Affiliation(s)
- Tong-Zhou Liang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China
| | - Zhe-Yu Jin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China
| | - Yue-Jun Lin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China
| | - Zi-Yi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China
| | - Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China
| | - Jian-Kun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China.
| | - Fan Yang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, the Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong, China.
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Sha Tin, 999077, Hong Kong, China.
| |
Collapse
|
6
|
Grajales-Reyes JG. Advances in energy balance & metabolism circuitry. ADVANCES IN GENETICS 2025; 113:1-28. [PMID: 40409794 DOI: 10.1016/bs.adgen.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Advancements in informatics, genetics, and neuroscience have greatly expanded our understanding of how the central nervous system (CNS) regulates energy balance and metabolism. This chapter explores the key neural circuits within the hypothalamus and brainstem that integrate behavioral and physiological processes to maintain metabolic homeostasis. It also examines the dynamic interplay between the CNS and peripheral organs, mediated through hormonal and neuronal signals, which fine-tune appetite, energy expenditure, and body weight. Furthermore, we highlight groundbreaking research that unveils molecular and cellular pathways governing energy regulation, representing a new frontier in addressing obesity and metabolic disorders. Innovative approaches, such as neurogenetic and neuromodulation techniques, are explored as promising strategies for improving weight management and metabolic health. By providing a comprehensive perspective on the mechanisms underlying energy balance, this chapter underscores the transformative potential of emerging therapeutic innovations.
Collapse
Affiliation(s)
- Jose G Grajales-Reyes
- Department of Anesthesiology, Yale University, New Haven, CT, United States; Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
7
|
Au E, Panganiban KJ, Wu S, Sun K, Humber B, Remington G, Agarwal SM, Giacca A, Pereira S, Hahn M. Antipsychotic-Induced Dysregulation of Glucose Metabolism Through the Central Nervous System: A Scoping Review of Animal Models. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025; 10:244-257. [PMID: 39461717 DOI: 10.1016/j.bpsc.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The use of antipsychotic drugs is associated with adverse metabolic effects. Disruptions in glucose metabolism such as hyperglycemia and insulin resistance have been shown to occur with antipsychotic use, independent of changes in body weight or adiposity. The regulation of whole-body glucose metabolism is partly mediated by the central nervous system. In particular, the hypothalamus and brainstem are responsive to peripheral energy signals and subsequently mediate feedback mechanisms to maintain peripheral glucose homeostasis. In this scoping review of preclinical in vivo studies, we aimed to explore central mechanisms through which antipsychotics dysregulate glucose metabolism. A systematic search for animal studies identified 29 studies that met our eligibility criteria for qualitative synthesis. The studies suggest that antipsychotic-induced changes in autonomic nervous system activity, certain neurotransmitter systems, expression of neuropeptides, and central insulin action mediate impairments in glucose metabolism. These findings provide insight into potential targets for the mitigation of the adverse effects of antipsychotics on glucose metabolism.
Collapse
Affiliation(s)
- Emily Au
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Kristoffer J Panganiban
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Sally Wu
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Kira Sun
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Bailey Humber
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Gary Remington
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Sri Mahavir Agarwal
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Adria Giacca
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Margaret Hahn
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Li Y, Mao J, Chai G, Zheng R, Liu X, Xie J. Neurobiological mechanisms of nicotine's effects on feeding and body weight. Neurosci Biobehav Rev 2025; 169:106021. [PMID: 39826824 DOI: 10.1016/j.neubiorev.2025.106021] [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: 11/29/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Nicotine, a neuroactive substance in tobacco products, has been widely studied for its effects on feeding and body weight, mostly focusing on the involvement of nervous system, metabolism, hormones, and gut microbiota. To elucidate the action mechanism of nicotine on feeding and body weight, especially the underlying neurobiological mechanisms, we reviewed the studies on nicotine's effects on feeding and body weight by the regulation of various nerve systems, energy expenditure, peripheral hormones, gut microbiota, etc. The role of neuronal signaling molecules such as AMP-activated protein kinase (AMPK) and kappa opioid receptor (κOR) were specialized in the nicotine-regulating energy expenditure. The energy homeostasis-related neurons, pro-opiomelanocortin (POMC), agouti-related peptide (AgRP), prolactin-releasing hormone (Prlh), etc, were discussed about the responsibility for nicotine's effects on feeding. Nicotine's actions on hypothalamus and its related neural circuits were described in view of peripheral nervous system, reward system, adipose browning, hormone secretion, and gut-brain axis. Elucidation of neurobiological mechanism of nicotine's actions on feeding and body weight will be of immense value to the therapeutic strategies of smoking, and advance the medicine research for the therapy of obesity.
Collapse
Affiliation(s)
- Ying Li
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China; Beijing Life Science Academy, Beijing, China
| | - Jian Mao
- Beijing Life Science Academy, Beijing, China
| | - Guobi Chai
- Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruimao Zheng
- Department of Anatomy Histology and Embryology School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xingyu Liu
- Beijing Life Science Academy, Beijing, China.
| | - Jianping Xie
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China; Beijing Life Science Academy, Beijing, China.
| |
Collapse
|
9
|
Wang B, Tang N, Chen S, Zhang X, Chen D, Li Z, Zhou B. Exploration of Appetite Regulation in Yangtze Sturgeon ( Acipenser dabryanus) During Weaning. Int J Mol Sci 2025; 26:950. [PMID: 39940719 PMCID: PMC11817240 DOI: 10.3390/ijms26030950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Yangtze sturgeon is an endangered fish species. After weaning, some Yangtze sturgeon fry refuse to consume any food, which causes a low survival rate during the artificial breeding period. This study showed that the body length and body weight of failed weaning Yangtze sturgeons were significantly lower than those of successful weaning sturgeons. Since the brain is the center of appetite regulation, RNA-seq of the brain was employed to analyze the differentially expressed genes and their biological functions in successfully and unsuccessfully weaned fry. After that, 82,151 unigenes and 3222 DEGs were obtained. Based on the results of RNA-seq, appetite factors, including POMC, CART, NPY and AgRP, were cloned, and then a weaning experiment was designed to explore the changes in appetite after feeding a microcapsule diet (weaning group). The results showed that, during the weaning period, the expression of CART was increased on the 1st and 3rd days but decreased on the 5th, 6th, 8th and 10th days. The expression of AgRP was downregulated on the 1st and 3rd days but upregulated on the 5th, 6th, 8th and 10th days. These findings indicate that appetite was suppressed in the early and middle periods but enhanced in the latter period of weaning and that CART may play an important role in the appetite-suppressing effect.
Collapse
Affiliation(s)
- Bin Wang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China; (B.W.); (N.T.)
- Fish Resources and Environment in the Upper Reaches of the Yangtze River Obervation and Research Station of Sichuan Province, Yibin 644000, China
| | - Ni Tang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China; (B.W.); (N.T.)
- Fish Resources and Environment in the Upper Reaches of the Yangtze River Obervation and Research Station of Sichuan Province, Yibin 644000, China
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (S.C.); (X.Z.); (D.C.)
| | - Shuhuang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (S.C.); (X.Z.); (D.C.)
| | - Xin Zhang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (S.C.); (X.Z.); (D.C.)
| | - Defang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (S.C.); (X.Z.); (D.C.)
| | - Zhiqiong Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (S.C.); (X.Z.); (D.C.)
| | - Bo Zhou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China; (B.W.); (N.T.)
- Fish Resources and Environment in the Upper Reaches of the Yangtze River Obervation and Research Station of Sichuan Province, Yibin 644000, China
| |
Collapse
|
10
|
Rivi V, Batabyal A, Benatti C, Tascedda F, Blom JMC, Lukowiak K. Quercetin, the new stress buster: Investigating the transcriptional and behavioral effects of this flavonoid on multiple stressors using Lymnaea stagnalis. Comp Biochem Physiol C Toxicol Pharmacol 2025; 287:110053. [PMID: 39442780 DOI: 10.1016/j.cbpc.2024.110053] [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: 09/05/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Growing evidence suggests that a flavonoid-rich diet can prevent or reverse the effects of stressors, although the underlying mechanisms remain poorly understood. One common and abundant flavonoid found in numerous foods is quercetin. This study utilizes the pond snail Lymnaea stagnalis, a valid model organism for learning and memory, and a simple but robust learning paradigm-operant conditioning of aerial respiration-to explore the behavioral and transcriptional effects of different stressors on snails' cognitive functions and to investigate whether quercetin exposure can prevent stress effects on learning and memory formation. Our findings demonstrate that three different stressors-severe food deprivation, lipopolysaccharide injection (an inflammatory challenge), and fluoride exposure (a neurotoxic agent)-block memory formation for operant conditioning and affect the expression levels of key targets related to stress response, energy balance, and immune response in the snails' central ring ganglia. Remarkably, exposing snails to quercetin for 1 h before stress presentation prevents these effects at both the behavioral and transcriptional levels, demonstrating the potent stress-preventive properties of quercetin. Despite the evolutionary distance from humans, L. stagnalis has proven to be a valuable model for studying conserved mechanisms by which bioactive compounds like quercetin mitigate the adverse effects of various stressors on cognitive functions across species. Moreover, these findings offer insights into quercetin's potential for mitigating stress-induced physiological and cognitive impairments.
Collapse
Affiliation(s)
- Veronica Rivi
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Anuradha Batabyal
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada; Department of Physical and Natural Sciences, FLAME University, Pune, India
| | - Cristina Benatti
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Tascedda
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy; CIB, Consorzio Interuniversitario Biotecnologie, Trieste, Italy; Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna Maria Catharina Blom
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Ken Lukowiak
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| |
Collapse
|
11
|
Espinal Abreu V, Barnes R, Borra V, Schurdak J, Perez-Tilve D. Chemogenetic engagement of different GPCR signaling pathways segregates the orexigenic activity from the control of whole-body glucose metabolism by AGRP neurons. Mol Metab 2025; 91:102079. [PMID: 39643082 PMCID: PMC11699438 DOI: 10.1016/j.molmet.2024.102079] [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: 10/09/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024] Open
Abstract
OBJECTIVE The control of energy balance involves neural circuits in the central nervous system, including AGRP neurons in the arcuate nucleus of the hypothalamus (ARC). AGRP neurons are crucial for energy balance and their increased activity during fasting is critical to promote feeding behavior. The activity of these neurons is influenced by multiple signals including those acting on G-protein coupled receptors (GPCR) activating different intracellular signaling pathways. We sought to determine whether discrete G-protein mediated signaling in AGRP neurons, promotes differential regulation of feeding and whole-body glucose homeostasis. METHODS To test the contribution of Gαq/11 or Gαs signaling, we developed congenital mouse lines expressing the different DREADD receptors (i.e., hM3q and rM3s), in AGRP neurons. Then we elicited chemogenetic activation of AGRP neurons in these mice during the postprandial state to determine the impact on feeding and glucose homeostasis. RESULTS Activation of AGRP neurons via hM3q and rM3s promoted hyperphagia. In contrast, only hM3q activation of AGRP neurons of the hypothalamic arcuate nucleus during the postprandial state enhanced whole-body glucose disposal by reducing sympathetic nervous system activity to the pancreas and liver, promoting glucose-stimulated insulin secretion, glycogen deposition and improving glucose tolerance. CONCLUSIONS These data indicate that AGRP neurons regulate food intake and glucose homeostasis through distinct GPCR-dependent signaling pathways and suggest that the transient increase in AGRP neuron activity may contribute to the beneficial effects of fasting on glycemic control.
Collapse
Affiliation(s)
- Valerie Espinal Abreu
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, USA
| | - Rachel Barnes
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, USA
| | - Vishnupriya Borra
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, USA
| | - Jennifer Schurdak
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, USA
| | - Diego Perez-Tilve
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, USA.
| |
Collapse
|
12
|
Webster AN, Becker JJ, Li C, Schwalbe DC, Kerspern D, Karolczak EO, Bundon CB, Onoharigho RA, Crook M, Jalil M, Godschall EN, Dame EG, Dawer A, Belmont-Rausch DM, Pers TH, Lutas A, Habib N, Güler AD, Krashes MJ, Campbell JN. Molecular connectomics reveals a glucagon-like peptide 1-sensitive neural circuit for satiety. Nat Metab 2024; 6:2354-2373. [PMID: 39627618 DOI: 10.1038/s42255-024-01168-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 10/17/2024] [Indexed: 12/11/2024]
Abstract
Liraglutide and other glucagon-like peptide 1 receptor agonists (GLP-1RAs) are effective weight loss drugs, but how they suppress appetite remains unclear. One potential mechanism is by activating neurons that inhibit the hunger-promoting Agouti-related peptide (AgRP) neurons of the arcuate hypothalamus (Arc). To identify these afferents, we developed a method combining rabies-based connectomics with single-nucleus transcriptomics. Here, we identify at least 21 afferent subtypes of AgRP neurons in the mouse mediobasal and paraventricular hypothalamus, which are predicted by our method. Among these are thyrotropin-releasing hormone (TRH)+ Arc (TRHArc) neurons, inhibitory neurons that express the Glp1r gene and are activated by the GLP-1RA liraglutide. Activating TRHArc neurons inhibits AgRP neurons and feeding, probably in an AgRP neuron-dependent manner. Silencing TRHArc neurons causes overeating and weight gain and attenuates liraglutide's effect on body weight. Our results demonstrate a widely applicable method for molecular connectomics, comprehensively identify local inputs to AgRP neurons and reveal a circuit through which GLP-1RAs suppress appetite.
Collapse
Affiliation(s)
- Addison N Webster
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
| | - Jordan J Becker
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Chia Li
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Dana C Schwalbe
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Damien Kerspern
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Eva O Karolczak
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | | | | | - Maisie Crook
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Maira Jalil
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | | | - Emily G Dame
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Adam Dawer
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | | | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Andrew Lutas
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Naomi Habib
- Center for Brain Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ali D Güler
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Michael J Krashes
- Section on Motivational Processes Underlying Appetite, Diabetes, Endocrinology & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
| | - John N Campbell
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA.
- Department of Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
13
|
Webster AN, Becker JJ, Li C, Schwalbe DC, Kerspern D, Karolczak EO, Bundon C, Onoharigho RA, Crook M, Jalil M, Godschall EN, Dame EG, Dawer A, Belmont-Rausch DM, Pers TH, Lutas A, Habib N, Guler AD, Krashes MJ, Campbell JN. Molecular Connectomics Reveals a Glucagon-Like Peptide 1 Sensitive Neural Circuit for Satiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.31.564990. [PMID: 37961449 PMCID: PMC10635031 DOI: 10.1101/2023.10.31.564990] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Liraglutide and other agonists of the glucagon-like peptide 1 receptor (GLP-1RAs) are effective weight loss drugs, but how they suppress appetite remains unclear. One potential mechanism is by activating neurons which inhibit hunger-promoting Agouti-related peptide (AgRP) neurons of the arcuate hypothalamus (Arc). To identify these afferents, we developed a method combining rabies-based connectomics with single-nuclei transcriptomics. Applying this method to AgRP neurons predicted at least 21 afferent subtypes in the mouse mediobasal and paraventricular hypothalamus. Among these are Trh+ Arc neurons, inhibitory neurons which express the Glp1r gene and are activated by the GLP-1RA liraglutide. Activating Trh+ Arc neurons inhibits AgRP neurons and feeding in an AgRP neuron-dependent manner. Silencing Trh+ Arc neurons causes over-eating and weight gain and attenuates liraglutide's effect on body weight. Our results demonstrate a widely applicable method for molecular connectomics, comprehensively identify local inputs to AgRP neurons, and reveal a circuit through which GLP-1RAs suppress appetite.
Collapse
|
14
|
Dodt S, Widdershooven NV, Dreisow ML, Weiher L, Steuernagel L, Wunderlich FT, Brüning JC, Fenselau H. NPY-mediated synaptic plasticity in the extended amygdala prioritizes feeding during starvation. Nat Commun 2024; 15:5439. [PMID: 38937485 PMCID: PMC11211344 DOI: 10.1038/s41467-024-49766-0] [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: 08/30/2023] [Accepted: 06/18/2024] [Indexed: 06/29/2024] Open
Abstract
Efficient control of feeding behavior requires the coordinated adjustment of complex motivational and affective neurocircuits. Neuropeptides from energy-sensing hypothalamic neurons are potent feeding modulators, but how these endogenous signals shape relevant circuits remains unclear. Here, we examine how the orexigenic neuropeptide Y (NPY) adapts GABAergic inputs to the bed nucleus of the stria terminalis (BNST). We find that fasting increases synaptic connectivity between agouti-related peptide (AgRP)-expressing 'hunger' and BNST neurons, a circuit that promotes feeding. In contrast, GABAergic input from the central amygdala (CeA), an extended amygdala circuit that decreases feeding, is reduced. Activating NPY-expressing AgRP neurons evokes these synaptic adaptations, which are absent in NPY-deficient mice. Moreover, fasting diminishes the ability of CeA projections in the BNST to suppress food intake, and NPY-deficient mice fail to decrease anxiety in order to promote feeding. Thus, AgRP neurons drive input-specific synaptic plasticity, enabling a selective shift in hunger and anxiety signaling during starvation through NPY.
Collapse
Affiliation(s)
- 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, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Noah V Widdershooven
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Marie-Luise Dreisow
- 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
| | - Lisa Weiher
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, 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
- Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, 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.
- Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931, Cologne, Germany.
| | - 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
|
15
|
Leeson-Payne A, Iyinikkel J, Malcolm C, Lam BYH, Sommer N, Dowsett GKC, Martinez de Morentin PB, Thompson D, Mackenzie A, Chianese R, Kentistou K, Gardner EJ, Perry JRB, Grassmann F, Speakman JR, Rochford JJ, Yeo GSH, Murray F, Heisler LK. Loss of GPR75 protects against non-alcoholic fatty liver disease and body fat accumulation. Cell Metab 2024; 36:1076-1087.e4. [PMID: 38653246 DOI: 10.1016/j.cmet.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/04/2023] [Accepted: 03/29/2024] [Indexed: 04/25/2024]
Abstract
Approximately 1 in 4 people worldwide have non-alcoholic fatty liver disease (NAFLD); however, there are currently no medications to treat this condition. This study investigated the role of adiposity-associated orphan G protein-coupled receptor 75 (GPR75) in liver lipid accumulation. We profiled Gpr75 expression and report that it is most abundant in the brain. Next, we generated the first single-cell-level analysis of Gpr75 and identified a subpopulation co-expressed with key appetite-regulating hypothalamic neurons. CRISPR-Cas9-deleted Gpr75 mice fed a palatable western diet high in fat adjusted caloric intake to remain in energy balance, thereby preventing NAFLD. Consistent with mouse results, analysis of whole-exome sequencing data from 428,719 individuals (UK Biobank) revealed that variants in GPR75 are associated with a reduced likelihood of hepatic steatosis. Here, we provide a significant advance in understanding of the expression and function of GPR75, demonstrating that it is a promising pharmaceutical target for NAFLD treatment.
Collapse
Affiliation(s)
| | - Jean Iyinikkel
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Cameron Malcolm
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Brian Y H Lam
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Nadine Sommer
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Georgina K C Dowsett
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | | | - Dawn Thompson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | - Katherine Kentistou
- Medical Research Council Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eugene J Gardner
- Medical Research Council Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John R B Perry
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Medical Research Council Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Felix Grassmann
- Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - John R Speakman
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Giles S H Yeo
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Fiona Murray
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| | - Lora K Heisler
- The Rowett Institute, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
16
|
Diao S, Chen C, Benani A, Magnan C, Van Steenwinckel J, Gressens P, Cruciani-Guglielmacci C, Jacquens A, Bokobza C. Preterm birth: A neuroinflammatory origin for metabolic diseases? Brain Behav Immun Health 2024; 37:100745. [PMID: 38511150 PMCID: PMC10950814 DOI: 10.1016/j.bbih.2024.100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/16/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Preterm birth and its related complications have become more and more common as neonatal medicine advances. The concept of "developmental origins of health and disease" has raised awareness of adverse perinatal events in the development of diseases later in life. To explore this concept, we propose that encephalopathy of prematurity (EoP) as a potential pro-inflammatory early life event becomes a novel risk factor for metabolic diseases in children/adolescents and adulthood. Here, we review epidemiological evidence that links preterm birth to metabolic diseases and discuss possible synergic roles of preterm birth and neuroinflammation from EoP in the development of metabolic diseases. In addition, we explore theoretical underlying mechanisms regarding developmental programming of the energy control system and HPA axis.
Collapse
Affiliation(s)
- Sihao Diao
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | | | | | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | | | - Alice Jacquens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Anesthesia and Critical Care, APHP-Sorbonne University, Hôpital La Pitié- Salpêtrière, Paris, France
| | - Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| |
Collapse
|
17
|
Guan D, Men Y, Bartlett A, Hernández MAS, Xu J, Yi X, Li HS, Kong D, Mazitschek R, Ozcan U. Central inhibition of HDAC6 re-sensitizes leptin signaling during obesity to induce profound weight loss. Cell Metab 2024; 36:857-876.e10. [PMID: 38569472 DOI: 10.1016/j.cmet.2024.02.007] [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: 06/22/2023] [Revised: 11/02/2023] [Accepted: 02/13/2024] [Indexed: 04/05/2024]
Abstract
Leptin resistance during excess weight gain significantly contributes to the recidivism of obesity to leptin-based pharmacological therapies. The mechanisms underlying the inhibition of leptin receptor (LepR) signaling during obesity are still elusive. Here, we report that histone deacetylase 6 (HDAC6) interacts with LepR, reducing the latter's activity, and that pharmacological inhibition of HDAC6 activity disrupts this interaction and augments leptin signaling. Treatment of diet-induced obese mice with blood-brain barrier (BBB)-permeable HDAC6 inhibitors profoundly reduces food intake and leads to potent weight loss without affecting the muscle mass. Genetic depletion of Hdac6 in Agouti-related protein (AgRP)-expressing neurons or administration with BBB-impermeable HDAC6 inhibitors results in a lack of such anti-obesity effect. Together, these findings represent the first report describing a mechanistically validated and pharmaceutically tractable therapeutic approach to directly increase LepR activity as well as identifying centrally but not peripherally acting HDAC6 inhibitors as potent leptin sensitizers and anti-obesity agents.
Collapse
Affiliation(s)
- Dongxian Guan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuqin Men
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander Bartlett
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jie Xu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xinchi Yi
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hu-Song Li
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Kong
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Mazitschek
- Massachusetts General Hospital, Center for Systems Biology, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Umut Ozcan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Zhang S, Zhang Y, Wen Z, Yang Y, Bu T, Wei R, Chen Y, Ni Q. Jinkui Shenqi pills ameliorate diabetes by regulating hypothalamic insulin resistance and POMC/AgRP expression and activity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155297. [PMID: 38342019 DOI: 10.1016/j.phymed.2023.155297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/10/2023] [Accepted: 12/16/2023] [Indexed: 02/13/2024]
Abstract
BACKGROUND Research on the imbalance of proopiomelanocortin (POMC)/agouti-related protein (AgRP) neurons in the hypothalamus holds potential insights into the pathophysiology of diabetes. Jinkui Shenqi pills (JSP), a prevalent traditional Chinese medicine, regulate hypothalamic function and treat diabetes. PURPOSE To investigate the hypoglycemic effect of JSP and explore the probable mechanism in treating diabetes. METHODS A type 2 diabetes mouse model was used to investigate the pharmacodynamics of JSP. The glucose-lowering efficacy of JSP was assessed through various metrics including body weight, food consumption, fasting blood glucose (FBG), serum insulin levels, and an oral glucose tolerance test (OGTT). To elucidate the modulatory effects of JSP on hypothalamic mechanisms, we quantified the expression and activity of POMC and AgRP and assessed the insulin-mediated phosphoinositide 3-kinase (PI3K)/protein kinase A (AKT)/forkhead box O1 (FOXO1) pathway in diabetic mice via western blotting and immunohistochemistry. Additionally, primary hypothalamic neurons were exposed to high glucose and palmitic acid levels to induce insulin resistance, and the influence of JSP on POMC/AgRP protein expression and activation was evaluated by PI3K protein inhibition using western blotting and immunofluorescence. RESULTS Medium- and high-dose JSP treatment effectively inhibited appetite, resulting in a steady declining trend in body weight, FBG, and OGTT results in diabetic mice (p < 0.05). These JSP groups also had significantly increased insulin levels (p < 0.05). Importantly, the medium-dose group exhibited notable protection of hypothalamic neuronal and synaptic structures, leading to augmentation of dendritic length and branching (p < 0.05). Furthermore, low-, medium-, and high-dose JSP groups exhibited increased phosphorylated (p) INSR, PI3K, pPI3K, AKT, and pAKT expression, as well as decreased FOXO1 and increased pFOXO1 expression, indicating improved hypothalamic insulin resistance in diabetic mice (p < 0.05). Treatment with 10% JSP-enriched serum produced a marked elevation of both expression and activation of POMC (p < 0.05), with a concurrent reduction in AgRP expression and activation within primary hypothalamic neurons (p < 0.05). Intriguingly, these effects could be attributed to the regulatory dynamics of PI3K activity. CONCLUSION Our findings suggest that JSP can ameliorate diabetes by regulating POMC/AgRP expression and activity. The insulin-mediated PI3K/AKT/FOXO1 pathway plays an important regulatory role in this intricate process.
Collapse
Affiliation(s)
- Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yueying Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhige Wen
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanan Yang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Tianjie Bu
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Ruoyu Wei
- Department of Traditional Chinese Medicine, The Fifth Hospital of Xingtai, Hebei, 054000, China
| | - Yupeng Chen
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qing Ni
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
19
|
Hahn MK, Giacca A, Pereira S. In vivo techniques for assessment of insulin sensitivity and glucose metabolism. J Endocrinol 2024; 260:e230308. [PMID: 38198372 PMCID: PMC10895285 DOI: 10.1530/joe-23-0308] [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: 09/23/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Metabolic tests are vital to determine in vivo insulin sensitivity and glucose metabolism in preclinical models, usually rodents. Such tests include glucose tolerance tests, insulin tolerance tests, and glucose clamps. Although these tests are not standardized, there are general guidelines for their completion and analysis that are constantly being refined. In this review, we describe metabolic tests in rodents as well as factors to consider when designing and performing these tests.
Collapse
Affiliation(s)
- Margaret K Hahn
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
| | - Adria Giacca
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Abasubong KP, Jiang GZ, Guo HX, Wang X, Huang YY, Li XF, Yan-Zou D, Liu WB, Desouky HE. Effects of a high-fat and high-carbohydrate diet on appetite regulation and central AMPK in the hypothalamus of blunt snout bream (Megalobrama amblycephala). J Anim Physiol Anim Nutr (Berl) 2024; 108:480-492. [PMID: 38014877 DOI: 10.1111/jpn.13908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a sensor of cellular energy changes and controls food intake. This study investigates the effect of a high-calorie diet (high fat diet [HFD], high carbohydrate diet [HCD] and high energy diet [HED]) on appetite and central AMPK in blunt snout bream. In the present study, fish (average initial weight 45.84 ± 0.07 g) were fed the control, HFD, HCD and HED in four replicates for 12 weeks. At the end of the feeding trial, the result showed that body mass index, specific growth rate, feed efficiency ratio and feed intake were not affected (p > 0.05) by dietary treatment. However, fish fed the HFD obtained a significantly higher (p < 0.05) lipid productive value, lipid gain and lipid intake than those fed the control diet, but no significant difference was attributed to others. Also, a significantly higher (p < 0.05) energy intake content was found in fish-fed HFD, HCD and HED than those given the control diet. Long-term HFD and HCD feeding significantly increased (p < 0.05) plasma glucose, glycated serum protein, advanced glycation end product, insulin and leptin content levels than the control group. Moreover, a significantly lower (p < 0.05) complex 1, 2 and 3 content was found in fish-fed HFD and HCD than in the control, but no differences (p > 0.05) were attributed to those in HED. Fish-fed HED significantly upregulated (p < 0.05) hypothalamic ampα 1 and ampα 2 expression, whereas the opposite trend was observed in the hypothalamic mammalian target of rapamycin than those in HFD and HCD compared to the control. However, hypothalamic neuropeptide y, peroxisome proliferator-activated receptor α (pparα), acetyl-coa oxidase and carnitine palmitoyltransferase 1 were significantly upregulated (p < 0.05) in the HCD group, while the opposite was seen in cholecystokinin expression compared to those in the control group. Our findings indicated that the central AMPK signal pathway and appetite were modulated according to the diet's energy level to regulate nutritional status and maintain energy homoeostasis in fish.
Collapse
Affiliation(s)
- Kenneth Prudence Abasubong
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Guang-Zhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Hui-Xing Guo
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Xi Wang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Yang-Yang Huang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Xiang-Fei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Dong Yan-Zou
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Wen-Bin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Hesham Eed Desouky
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
- Department of Animal and Poultry Production, Faculty of Agriculture, Damanhour University, Damanhour, Egypt
| |
Collapse
|
21
|
De Solis AJ, Del Río-Martín A, Radermacher J, Chen W, Steuernagel L, Bauder CA, Eggersmann FR, Morgan DA, Cremer AL, Sué M, Germer M, Kukat C, Vollmar S, Backes H, Rahmouni K, Kloppenburg P, Brüning JC. Reciprocal activity of AgRP and POMC neurons governs coordinated control of feeding and metabolism. Nat Metab 2024; 6:473-493. [PMID: 38378998 DOI: 10.1038/s42255-024-00987-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Agouti-related peptide (AgRP)-expressing and proopiomelanocortin (POMC)-expressing neurons reciprocally regulate food intake. Here, we combine non-interacting recombinases to simultaneously express functionally opposing chemogenetic receptors in AgRP and POMC neurons for comparing metabolic responses in male and female mice with simultaneous activation of AgRP and inhibition of POMC neurons with isolated activation of AgRP neurons or isolated inhibition of POMC neurons. We show that food intake is regulated by the additive effect of AgRP neuron activation and POMC neuron inhibition, while systemic insulin sensitivity and gluconeogenesis are differentially modulated by isolated-versus-simultaneous regulation of AgRP and POMC neurons. We identify a neurocircuit engaging Npy1R-expressing neurons in the paraventricular nucleus of the hypothalamus, where activated AgRP neurons and inhibited POMC neurons cooperate to promote food consumption and activate Th+ neurons in the nucleus tractus solitarii. Collectively, these results unveil how food intake is precisely regulated by the simultaneous bidirectional interplay between AgRP and POMC neurocircuits.
Collapse
Affiliation(s)
- Alain J De Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Jan Radermacher
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Fynn R Eggersmann
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Anna-Lena Cremer
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Michael Sué
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Maximilian Germer
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Christian Kukat
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Stefan Vollmar
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Heiko Backes
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
22
|
Ali NH, Al‐Kuraishy HM, Al‐Gareeb AI, Alnaaim SA, Alexiou A, Papadakis M, Khalifa AA, Saad HM, Batiha GE. Neprilysin inhibitors and risk of Alzheimer's disease: A future perspective. J Cell Mol Med 2024; 28:e17993. [PMID: 37847125 PMCID: PMC10826440 DOI: 10.1111/jcmm.17993] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disease with multifaceted neuropathological disorders. AD is characterized by intracellular accumulation of phosphorylated tau proteins and extracellular deposition of amyloid beta (Aβ). Various protease enzymes, including neprilysin (NEP), are concerned with the degradation and clearance of Aβ. Indeed, a defective neuronal clearance pathway due to the dysfunction of degradation enzymes might be a possible mechanism for the accumulation of Aβ and subsequent progression of AD neuropathology. NEP is one of the most imperative metalloproteinase enzymes involved in the clearance of Aβ. This review aimed to highlight the possible role of NEP inhibitors in AD. The combination of sacubitril and valsartan which is called angiotensin receptor blocker and NEP inhibitor (ARNI) may produce beneficial and deleterious effects on AD neuropathology. NEP inhibitors might increase the risk of AD by the inhibition of Aβ clearance, and increase brain bradykinin (BK) and natriuretic peptides (NPs), which augment the pathogenesis of AD. These verdicts come from animal model studies, though they may not be applied to humans. However, clinical studies revealed promising safety findings regarding the use of ARNI. Moreover, NEP inhibition increases various neuroprotective peptides involved in inflammation, glucose homeostasis and nerve conduction. Also, NEP inhibitors may inhibit dipeptidyl peptidase 4 (DPP4) expression, ameliorating insulin and glucagon-like peptide 1 (GLP-1) levels. These findings proposed that NEP inhibitors may have a protective effect against AD development by increasing GLP-1, neuropeptide Y (NPY) and substance P, and deleterious effects by increasing brain BK. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Saud A. Alnaaim
- Clinical Neurosciences Department, College of MedicineKing Faisal UniversityHofufSaudi Arabia
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Asmaa A. Khalifa
- Department of Pharmacology and Therapeutics, Faculty of PharmacyPharos University in AlexandriaAlexandriaEgypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
23
|
Najm Al-Halboosi DA, Savchenko O, Heisler LK, Sylantyev S. Modulation of GABA release by 5-HT 1B receptors: An interplay with AMPA-receptors and voltage-gated Ca 2+ channels. Neuropharmacology 2023; 241:109758. [PMID: 37827445 DOI: 10.1016/j.neuropharm.2023.109758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Obesity has become a worldwide health challenge and commonly results from the intake of more calories than the body requires. The brain represents the master controller of food intake and as such has been the target of obesity medications. However, key mechanisms of druggable targets remain to be defined. Neurons within the arcuate nucleus of the hypothalamus co-expressing neuropeptide Y (NPY), agouti-related protein (AgRP) and GABA (NAG) are fundamental stimulators of hunger and food intake. NAG neurons also inhibit local satiety-promoting pro-opiomelanocortin (POMC) neurons. Agonists of the 1B subtype of metabotropic serotonin receptor (5-HT1BR) reduce food intake in part through the inhibition of hunger-promoting NAG neurons. We first confirmed that 5-HT1BR activation suppressed intake of a palatable Western diet in a mouse model of common dietary-induced obesity and genetically prone obesity. Next, we combined several electrophysiological approaches to analyse the effect of 5-HT1BRs in NAG neuron cell activity and GABA release. 5-HT1BR activation reduced NAG neuron action potential frequency and neurotransmitter release. We found that 5-HT1BR impact on GABA release from NAG neurons is mediated through voltage-gated Ca2+ channels with a critical input from glutamate receptors of AMPA subtype (AMPARs). As a fundamental outcome, this type of interplay provides an uncommon example of metabotropic action of AMPARs which regulates inhibitory signalling due to modulation of GABA release. As a translational outcome, our results provide a key mechanism through which 5-HT1BR drugs inhibit appetite-stimulating neurons within the brain to suppress food intake. This article is part of the Special Issue on "Ukrainian Neuroscience".
Collapse
Affiliation(s)
| | - Olena Savchenko
- National Technical University of Ukraine "Igor Sikorsky KPI", Department of Biotechnology, 37 Beresteiskyi Ave., Kyiv, 03056, Ukraine
| | - Lora K Heisler
- Rowett Institute, University of Aberdeen, Ashgrove Rd. West, Aberdeen, AB25 2ZD, UK
| | - Sergiy Sylantyev
- Rowett Institute, University of Aberdeen, Ashgrove Rd. West, Aberdeen, AB25 2ZD, UK; Odesa National Mechnikov University, Biological Department, 2 Shampansky Ln., Odesa, 65015, Ukraine.
| |
Collapse
|
24
|
Doan KV, Tran LT, Yang DJ, Ha TTA, Mai TD, Kim SK, DePinho RA, Shin DM, Choi YH, Kim KW. Astrocytic FoxO1 in the hypothalamus regulates metabolic homeostasis by coordinating neuropeptide Y neuron activity. Glia 2023; 71:2735-2752. [PMID: 37655904 DOI: 10.1002/glia.24448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 09/02/2023]
Abstract
The forkhead box transcription factor O1 (FoxO1) is expressed ubiquitously throughout the central nervous system, including in astrocytes, the most prevalent glial cell type in the brain. While the role of FoxO1 in hypothalamic neurons in controlling food intake and energy balance is well-established, the contribution of astrocytic FoxO1 in regulating energy homeostasis has not yet been determined. In the current study, we demonstrate the essential role of hypothalamic astrocytic FoxO1 in maintaining normal neuronal activity in the hypothalamus and whole-body glucose metabolism. Inhibition of FoxO1 function in hypothalamic astrocytes shifts the cellular metabolism from glycolysis to oxidative phosphorylation, enhancing astrocyte ATP production and release meanwhile decreasing astrocytic export of lactate. As a result, specific deletion of astrocytic FoxO1, particularly in the hypothalamus, causes a hyperactivation of hypothalamic neuropeptide Y neurons, which leads to an increase in acute feeding and impaired glucose regulation and ultimately results in diet-induced obesity and systemic glucose dyshomeostasis.
Collapse
Affiliation(s)
- Khanh Van Doan
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Le Trung Tran
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Dong Joo Yang
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Thu Thi Anh Ha
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Thi Dang Mai
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Seul Ki Kim
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dong-Min Shin
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Yun-Hee Choi
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Ki Woo Kim
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
- Department of Applied Life Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, Republic of Korea
| |
Collapse
|
25
|
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: 74] [Impact Index Per Article: 37.0] [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
|
26
|
Jaschke NP, Wang A. The neurocircuitry of fasting-induced glucocorticoid release. Cell Metab 2023; 35:1497-1499. [PMID: 37673035 DOI: 10.1016/j.cmet.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023]
Abstract
Activation of the hypothalamus-pituitary-adrenal gland (HPA) axis confers adaptations to homeostatic perturbations including food scarcity. A comprehensive new study by Douglass et al. disentangled how agouti-related peptide (AgRP)-expressing neurons of the arcuate nucleus (ARC) trigger rapid HPA-axis activation in response to fasting, which is mediated by repression of a tonic, inhibitory neuro circuit.
Collapse
Affiliation(s)
- Nikolai P Jaschke
- Division of Endocrinology, Department of Medicine III, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| | - Andrew Wang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
27
|
Sweeney P, Gimenez LE, Hernandez CC, Cone RD. Targeting the central melanocortin system for the treatment of metabolic disorders. Nat Rev Endocrinol 2023; 19:507-519. [PMID: 37365323 DOI: 10.1038/s41574-023-00855-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors.
Collapse
Affiliation(s)
- Patrick Sweeney
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular, Cellular, and Developmental Biology, College of Literature Science and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Chen WH, Shi YC, Huang QY, Chen JM, Wang ZY, Lin S, Shi QY. Potential for NPY receptor-related therapies for polycystic ovary syndrome: an updated review. Hormones (Athens) 2023; 22:441-451. [PMID: 37452264 PMCID: PMC10449684 DOI: 10.1007/s42000-023-00460-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine disease that can cause female infertility and bring economic burden to families and to society. The clinical and/or biochemical manifestations include hyperandrogenism, persistent anovulation, and polycystic ovarian changes, often accompanied by insulin resistance and obesity. Although its pathogenesis is unclear, PCOS involves the abnormal regulation of the hypothalamic-pituitary-ovarian axis and the abnormal activation of GnRH neurons. Neuropeptide Y (NPY) is widely distributed in the arcuate nucleus of the hypothalamus and functions as the physiological integrator of two neuroendocrine systems, one governing feeding and the other controlling reproduction. In recent years, an increasing number of studies have focused on the improvement of the reproductive and metabolic status of PCOS through the therapeutic application of NPY and its receptors. In this review, we summarize the central and peripheral regulation of NPY and its receptors in the development of PCOS and discuss the potential for NPY receptor-related therapies for PCOS.
Collapse
Affiliation(s)
- Wei-Hong Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Yan-Chuan Shi
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Qiao-Yi Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Jia-Ming Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Zhi-Yi Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
| | - Qi-Yang Shi
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
| |
Collapse
|
29
|
Ma HT, Zhang HC, Zuo ZF, Liu YX. Heterogeneous organization of Locus coeruleus: An intrinsic mechanism for functional complexity. Physiol Behav 2023; 268:114231. [PMID: 37172640 DOI: 10.1016/j.physbeh.2023.114231] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Locus coeruleus (LC) is a small nucleus located deep in the brainstem that contains the majority of central noradrenergic neurons, which provide the primary source of noradrenaline (NA) throughout the entire central nervous system (CNS).The release of neurotransmitter NA is considered to modulate arousal, sensory processing, attention, aversive and adaptive stress responses as well as high-order cognitive function and memory, with the highly ramified axonal arborizations of LC-NA neurons sending wide projections to the targeted brain areas. For over 30 years, LC was thought to be a homogeneous nucleus in structure and function due to the widespread uniform release of NA by LC-NA neurons and simultaneous action in several CNS regions, such as the prefrontal cortex, hippocampus, cerebellum, and spinal cord. However, recent advances in neuroscience tools have revealed that LC is probably not so homogeneous as we previous thought and exhibits heterogeneity in various aspects. Accumulating studies have shown that the functional complexity of LC may be attributed to its heterogeneity in developmental origin, projection patterns, topography distribution, morphology and molecular organization, electrophysiological properties and sex differences. This review will highlight the heterogeneity of LC and its critical role in modulating diverse behavioral outcomes.
Collapse
Affiliation(s)
- Hai-Tao Ma
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, 100069, China.
| | - Hao-Chen Zhang
- Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, 100069, China
| | - Zhong-Fu Zuo
- Department of Human Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121000, China
| | - Ying-Xue Liu
- Department of Human Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
30
|
Luigi-Sierra MG, Guan D, López-Béjar M, Casas E, Olvera-Maneu S, Gardela J, Palomo MJ, Osuagwuh UI, Ohaneje UL, Mármol-Sánchez E, Amills M. A protein-coding gene expression atlas from the brain of pregnant and non-pregnant goats. Front Genet 2023; 14:1114749. [PMID: 37519888 PMCID: PMC10382233 DOI: 10.3389/fgene.2023.1114749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Background: The brain is an extraordinarily complex organ with multiple anatomical structures involved in highly specialized functions related with behavior and physiological homeostasis. Our goal was to build an atlas of protein-coding gene expression in the goat brain by sequencing the transcriptomes of 12 brain regions in seven female Murciano-Granadina goats, from which three of them were 1-month pregnant. Results: Between 14,889 (cerebellar hemisphere) and 15,592 (pineal gland) protein-coding genes were expressed in goat brain regions, and most of them displayed ubiquitous or broad patterns of expression across tissues. Principal component analysis and hierarchical clustering based on the patterns of mRNA expression revealed that samples from certain brain regions tend to group according to their position in the anterior-posterior axis of the neural tube, i.e., hindbrain (pons and medulla oblongata), midbrain (rostral colliculus) and forebrain (frontal neocortex, olfactory bulb, hypothalamus, and hippocampus). Exceptions to this observation were cerebellum and glandular tissues (pineal gland and hypophysis), which showed highly divergent mRNA expression profiles. Differential expression analysis between pregnant and non-pregnant goats revealed moderate changes of mRNA expression in the frontal neocortex, hippocampus, adenohypophysis and pons, and very dramatic changes in the olfactory bulb. Many genes showing differential expression in this organ are related to olfactory function and behavior in humans. Conclusion: With the exception of cerebellum and glandular tissues, there is a relationship between the cellular origin of sampled regions along the anterior-posterior axis of the neural tube and their mRNA expression patterns in the goat adult brain. Gestation induces substantial changes in the mRNA expression of the olfactory bulb, a finding consistent with the key role of this anatomical structure on the development of maternal behavior.
Collapse
Affiliation(s)
| | - Dailu Guan
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Manel López-Béjar
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Encarna Casas
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Sergi Olvera-Maneu
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jaume Gardela
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - María Jesús Palomo
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Uchebuchi Ike Osuagwuh
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Uchechi Linda Ohaneje
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Emilio Mármol-Sánchez
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Marcel Amills
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
31
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [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: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
32
|
Maynard CW, Gilbert E, Yan F, Cline MA, Dridi S. Peripheral and Central Impact of Methionine Source and Level on Growth Performance, Circulating Methionine Levels and Metabolism in Broiler Chickens. Animals (Basel) 2023; 13:1961. [PMID: 37370471 DOI: 10.3390/ani13121961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The present study was designed to evaluate the effects of DL-methionine (DL-Met) 2-hydroxy-4-(methylthio) butanoic acid (HMTBa), or S-(5'-Adenosyl)-L-methionine chloride (SAM), using feeding trial and central administration, on live performance, plasma metabolites, and the expression of feeding-related hypothalamic neuropeptides in broilers raised to a market age (35 d). Final average body weight (BW) and feed conversion ratio (FCR) from the feeding trial exceeded the performance measurements published by the primary breeder. At d35, the MTBHa group had better BW and lower feed intake, which resulted in a better FCR than the DL-Met group at 87 TSAA to lysine. At the molecular levels, the expression of hypothalamic neuropeptide (NPY) and monocarboxylate transporter (MCT) 2 did not differ between all treated groups; however, the mRNA abundances of hypothalamic MCT1 and orexin (ORX) were significantly upregulated in DL-Met- treated groups compared to the control. The ICV administration of SAM significantly reduced feed intake at all tested periods (from 30 to 180 min post injection) compared to the aCSF-treated group (control). The central administration of HMTBa increased feed intake, which reached a significant level only 60 min post administration, compared to the control group. ICV administration of DL-Met slightly increased feed intake compared to the control group, but the difference was not statistically discernable. Quantitative real-time PCR analysis showed that the hypothalamic expression of NPY, cocaine- and amphetamine-regulated transcript, MCT1, and MCT2 was significantly upregulated in the ICV-HMTBa group compared to the aCSF birds. The hypothalamic expression of the mechanistic target of rapamycin (mTOR), AMP-activated protein kinase (AMPKα1), D-amino acid oxidase, and hydroxyacid oxidase was significantly upregulated in DL-Met compared to the control group. The mRNA abundances of ORX were significantly increased in the hypothalamus of both DL-Met and HMTBa groups compared to the aCSF birds; however, mTOR gene expression was significantly downregulated in the SAM compared to the control group. Taken together, these data show, for the first time, that DL-Met and HMTBa have a common downstream (ORX) pathway, but also a differential central pathway, typically NPY-MCT for HMTBa and mTOR-AMPK for methionine.
Collapse
Affiliation(s)
- Craig W Maynard
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Elizabeth Gilbert
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Frances Yan
- Novus International, Saint Charles, MO 63304, USA
| | - Mark A Cline
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
33
|
Chen W, Mehlkop O, Scharn A, Nolte H, Klemm P, Henschke S, Steuernagel L, Sotelo-Hitschfeld T, Kaya E, Wunderlich CM, Langer T, Kononenko NL, Giavalisco P, Brüning JC. Nutrient-sensing AgRP neurons relay control of liver autophagy during energy deprivation. Cell Metab 2023; 35:786-806.e13. [PMID: 37075752 PMCID: PMC10173804 DOI: 10.1016/j.cmet.2023.03.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/01/2023] [Accepted: 03/27/2023] [Indexed: 04/21/2023]
Abstract
Autophagy represents a key regulator of aging and metabolism in sensing energy deprivation. We find that fasting in mice activates autophagy in the liver paralleled by activation of hypothalamic AgRP neurons. Optogenetic and chemogenetic activation of AgRP neurons induces autophagy, alters phosphorylation of autophagy regulators, and promotes ketogenesis. AgRP neuron-dependent induction of liver autophagy relies on NPY release in the paraventricular nucleus of the hypothalamus (PVH) via presynaptic inhibition of NPY1R-expressing neurons to activate PVHCRH neurons. Conversely, inhibiting AgRP neurons during energy deprivation abrogates induction of hepatic autophagy and rewiring of metabolism. AgRP neuron activation increases circulating corticosterone concentrations, and reduction of hepatic glucocorticoid receptor expression attenuates AgRP neuron-dependent activation of hepatic autophagy. Collectively, our study reveals a fundamental regulatory principle of liver autophagy in control of metabolic adaptation during nutrient deprivation.
Collapse
Affiliation(s)
- Weiyi Chen
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Oliver Mehlkop
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Alexandra Scharn
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931 Cologne, Germany
| | - Paul Klemm
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Sinika Henschke
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Lukas Steuernagel
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Tamara Sotelo-Hitschfeld
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Ecem Kaya
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Claudia Maria Wunderlich
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931 Cologne, Germany
| | - Natalia L Kononenko
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Patrick Giavalisco
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931 Cologne, Germany
| | - Jens Claus Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
34
|
Wang X, Ge S, Zhang C. Bed nuclei of the stria terminalis: A key hub in the modulation of anxiety. Eur J Neurosci 2023; 57:900-917. [PMID: 36725691 DOI: 10.1111/ejn.15926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 01/12/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023]
Abstract
The bed nuclei of the stria terminalis (BST) is recognised as a pivotal integrative centre for monitoring emotional valence. It is implicated in the regulation of diverse affective states and motivated behaviours, and decades of research have firmly established its critical role in anxiety-related behavioural processes. Researchers have recently intricately dissected the BST's dynamic activities, its connection patterns and its functions with respect to specific cell types using multiple techniques such as optogenetics, in vivo calcium imaging and transgenic tools to unmask the complex circuitry mechanisms that underlie anxiety. In this review, we principally focus on studies of anxiety-involved neuromodulators within the BST and provide a comprehensive architecture of the anxiety network-highlighting the BST as a key hub in orchestrating anxiety-like behaviour. We posit that these promising efforts will contribute to the identification of an accurate roadmap for future treatment of anxiety disorders.
Collapse
Affiliation(s)
- Xinxin Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
35
|
Huang A, Maier MT, Vagena E, Xu AW. Modulation of foraging-like behaviors by cholesterol-FGF19 axis. Cell Biosci 2023; 13:20. [PMID: 36732847 PMCID: PMC9893607 DOI: 10.1186/s13578-023-00955-2] [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: 10/17/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Foraging for food precedes food consumption and is an important component of the overall metabolic programming that regulates feeding. Foraging is governed by central nervous system neuronal circuits but how it is influenced by diet and hormonal signals is still not well understood. RESULTS In this study, we show that dietary cholesterol exerted suppressive effects on locomotor activity and that these effects were partially mediated by the neuropeptide Agouti-related protein (AgRP). High dietary cholesterol stimulated intestinal expression of fibroblast growth factor 15 (Fgf15), an ortholog of the human fibroblast growth factor 19 (FGF19). Intracerebroventricular infusion of FGF19 peptide reduced exploratory activity in the open field test paradigm. On the other hand, the lack of dietary cholesterol enhanced exploratory activity in the open field test, but this effect was abolished by central administration of FGF19. CONCLUSIONS Experiments in this study show that dietary cholesterol suppresses locomotor activity and foraging-like behaviors, and this regulation is in part mediated by AgRP neurons. Dietary cholesterol or the central action of FGF19 suppresses exploratory behaviors, and the anxiogenic effects of dietary cholesterol may be mediated by the effect of FGF19 in the mouse brain. This study suggests that dietary cholesterol and intestinal hormone FGF15/19 signal a satiating state to the brain, thereby suppressing foraging-like behaviors.
Collapse
Affiliation(s)
- Alyssa Huang
- Diabetes Center, University of California, San Francisco, CA, 94143, USA
| | - Matthew T Maier
- Diabetes Center, University of California, San Francisco, CA, 94143, USA
| | - Eirini Vagena
- Diabetes Center, University of California, San Francisco, CA, 94143, USA
| | - Allison W Xu
- Diabetes Center, University of California, San Francisco, CA, 94143, USA. .,Department of Anatomy, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
36
|
Sharma S, Littman R, Tompkins J, Arneson D, Contreras J, Dajani AH, Ang K, Tsanhani A, Sun X, Jay PY, Herzog H, Yang X, Ajijola OA. Tiered Sympathetic Control of Cardiac Function Revealed by Viral Tracing and Single Cell Transcriptome Profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524575. [PMID: 36711942 PMCID: PMC9882306 DOI: 10.1101/2023.01.18.524575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The cell bodies of postganglionic sympathetic neurons innervating the heart primarily reside in the stellate ganglion (SG), alongside neurons innervating other organs and tissues. Whether cardiac-innervating stellate ganglionic neurons (SGNs) exhibit diversity and distinction from those innervating other tissues is not known. To identify and resolve the transcriptomic profiles of SGNs innervating the heart we leveraged retrograde tracing techniques using adeno-associated virus (AAV) expressing fluorescent proteins (GFP or Td-tomato) with single cell RNA sequencing. We investigated electrophysiologic, morphologic, and physiologic roles for subsets of cardiac-specific neurons and found that three of five adrenergic SGN subtypes innervate the heart. These three subtypes stratify into two subpopulations; high (NA1a) and low (NA1b and NA1c) Npy-expressing cells, exhibit distinct morphological, neurochemical, and electrophysiologic characteristics. In physiologic studies in transgenic mouse models modulating NPY signaling, we identified differential control of cardiac responses by these two subpopulations to high and low stress states. These findings provide novel insights into the unique properties of neurons responsible for cardiac sympathetic regulation, with implications for novel strategies to target specific neuronal subtypes for sympathetic blockade in cardiac disease.
Collapse
|
37
|
Jia X, Chen S, Li X, Tao S, Lai J, Liu H, Huang K, Tian Y, Wei P, Yang F, Lu Z, Chen Z, Liu XA, Xu F, Wang L. Divergent neurocircuitry dissociates two components of the stress response: glucose mobilization and anxiety-like behavior. Cell Rep 2022; 41:111586. [DOI: 10.1016/j.celrep.2022.111586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/19/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022] Open
|
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: 15] [Impact Index Per Article: 5.0] [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
|
Si R, Pan D, Wang Z, Chen Y, Cao J. Regulation of the central melanocortin system on energy balance in mammals and birds. Neuropeptides 2022; 95:102267. [PMID: 35752067 DOI: 10.1016/j.npep.2022.102267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022]
Abstract
Agouti-related protein/neuropeptide Y (AgRP/NPY) neurons promote feeding, while proopiomelanocortin/cocaine- and amphetamine-regulated transcript (POMC/CART) neurons and melanocortin receptor neurons inhibit feeding; these three types of neurons play vital roles in regulating feeding. The central melanocortin system composed of these neurons is critical for the regulation of food intake and energy metabolism. It regulates energy intake and consumption by activating or inhibiting the activities of AgRP/NPY neurons and POMC/CART neurons and then affects the feeding behaviour of animals to maintain the energy balance. Meanwhile, organisms can also positively or negatively regulate energy homeostasis through the negative feedback of the neuron system. With further studies, understanding of the process and factors involved in the energy balance regulation of mammals and birds can be improved, which will provide a favourable scientific basis to reduce costs and improve meat production in production and breeding.
Collapse
Affiliation(s)
- Rongrong Si
- Laboratory of Anatomy of Domestic Animals, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Deng Pan
- Laboratory of Anatomy of Domestic Animals, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China.
| |
Collapse
|
40
|
Abstract
Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) are essential to normal growth, metabolism, and body composition, but in acromegaly, excesses of these hormones strikingly alter them. In recent years, the use of modern methodologies to assess body composition in patients with acromegaly has revealed novel aspects of the acromegaly phenotype. In particular, acromegaly presents a unique pattern of body composition changes in the setting of insulin resistance that we propose herein to be considered an acromegaly-specific lipodystrophy. The lipodystrophy, initiated by a distinctive GH-driven adipose tissue dysregulation, features insulin resistance in the setting of reduced visceral adipose tissue (VAT) mass and intra-hepatic lipid (IHL) but with lipid redistribution, resulting in ectopic lipid deposition in muscle. With recovery of the lipodystrophy, adipose tissue mass, especially that of VAT and IHL, rises, but insulin resistance is lessened. Abnormalities of adipose tissue adipokines may play a role in the disordered adipose tissue metabolism and insulin resistance of the lipodystrophy. The orexigenic hormone ghrelin and peptide Agouti-related peptide may also be affected by active acromegaly as well as variably by acromegaly therapies, which may contribute to the lipodystrophy. Understanding the pathophysiology of the lipodystrophy and how acromegaly therapies differentially reverse its features may be important to optimizing the long-term outcome for patients with this disease. This perspective describes evidence in support of this acromegaly lipodystrophy model and its relevance to acromegaly pathophysiology and the treatment of patients with acromegaly.
Collapse
Affiliation(s)
- Pamela U. Freda
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
41
|
Cho JH, Kim K, Cho HC, Lee J, Kim EK. Silencing of hypothalamic FGF11 prevents diet-induced obesity. Mol Brain 2022; 15:75. [PMID: 36064426 PMCID: PMC9447329 DOI: 10.1186/s13041-022-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/21/2022] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor 11 (FGF11) is a member of the intracellular fibroblast growth factor family. Here, we report the central role of FGF11 in the regulation of metabolism. Lentiviral injection of Fgf11 shRNA into the arcuate nucleus of the mouse hypothalamus decreased weight gain and fat mass, increased brown adipose tissue thermogenesis, and improved glucose and insulin intolerances under high-fat diet conditions. Fgf11 was expressed in the NPY–expressing neurons, and Fgf11 knockdown considerably decreased Npy expression and projection, leading to increased expression of tyrosine hydroxylase in the paraventricular nucleus. Mechanistically, FGF11 regulated Npy gene expression through the glycogen synthase kinase 3–cAMP response element-binding protein pathway. Our study defines the physiological significance of hypothalamic FGF11 in the regulation of metabolism in response to overnutrition such as high-fat diet.
Collapse
Affiliation(s)
- Jae Hyun Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Kyungchan Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Han Chae Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Jaemeun Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea. .,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea.
| |
Collapse
|
42
|
Yao Y, Hu Y, Yang J, Zhang C, He Y, Qi H, Zeng Y, Zhang A, Liu X, Zhu X. Inhibition of neuronal nitric oxide synthase protects against hippocampal neuronal injuries by increasing neuropeptide Y expression in temporal lobe epilepsy mice. Free Radic Biol Med 2022; 188:45-61. [PMID: 35714846 DOI: 10.1016/j.freeradbiomed.2022.06.221] [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: 01/11/2022] [Revised: 04/23/2022] [Accepted: 06/11/2022] [Indexed: 10/18/2022]
Abstract
Neuronal nitric oxide synthase (nNOS) plays a pivotal role in the pathological process of neuronal injury in the development of epilepsy. Our previous study has demonstrated that nitric oxide (NO) derived from nNOS in the epileptic brain is neurotoxic due to its reaction with the superoxide radical with the formation of peroxynitrite. Neuropeptide Y (NPY) is widely expressed in the mammalian brain, which has been implicated in energy homeostasis and neuroprotection. Recent studies suggest that nNOS may act as a mediator of NPY signaling. Here in this study, we sought to determine whether NPY expression is regulated by nNOS, and if so, whether the regulation of NPY by nNOS is associated with the neuronal injuries in the hippocampus of epileptic brain. Our results showed that pilocarpine-induced temporal lobe epilepsy (TLE) mice exhibited an increased level of nNOS expression and a decreased level of NPY expression along with hippocampal neuronal injuries and cognition deficit. Genetic deletion of nNOS gene, however, significantly upregulated hippocampal NPY expression and reduced TLE-induced hippocampal neuronal injuries and cognition decline. Knockdown of NPY abolished nNOS depletion-induced neuroprotection and cognitive improvement in the TLE mice, suggesting that inhibition of nNOS protects against hippocampal neuronal injuries by increasing neuropeptide Y expression in TLE mice. Targeting nNOS-NPY signaling pathway in the epileptic brain might provide clinical benefit by attenuating neuronal injuries and preventing cognitive deficits in epilepsy patients.
Collapse
Affiliation(s)
- Yuanyuan Yao
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Yang Hu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Jiurong Yang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Canyu Zhang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Yuqi He
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Honggang Qi
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Yu Zeng
- National Residents Clinical Skills Training Center, Medical School of Southeast University, Nanjing, China
| | - Aifeng Zhang
- Department of Pathology, Medical School of Southeast University, Nanjing, China
| | - Xiufang Liu
- Department of Pathogenic Biology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Xinjian Zhu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
43
|
Alcantara IC, Tapia APM, Aponte Y, Krashes MJ. Acts of appetite: neural circuits governing the appetitive, consummatory, and terminating phases of feeding. Nat Metab 2022; 4:836-847. [PMID: 35879462 PMCID: PMC10852214 DOI: 10.1038/s42255-022-00611-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
The overconsumption of highly caloric and palatable foods has caused a surge in obesity rates in the past half century, thereby posing a healthcare challenge due to the array of comorbidities linked to heightened body fat accrual. Developing treatments to manage body weight requires a grasp of the neurobiological basis of appetite. In this Review, we discuss advances in neuroscience that have identified brain regions and neural circuits that coordinate distinct phases of eating: food procurement, food consumption, and meal termination. While pioneering work identified several hypothalamic nuclei to be involved in feeding, more recent studies have explored how neuronal populations beyond the hypothalamus, such as the mesolimbic pathway and nodes in the hindbrain, interconnect to modulate appetite. We also examine how long-term exposure to a calorically dense diet rewires feeding circuits and alters the response of motivational systems to food. Understanding how the nervous system regulates eating behaviour will bolster the development of medical strategies that will help individuals to maintain a healthy body weight.
Collapse
Affiliation(s)
- Ivan C Alcantara
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | | | - Yeka Aponte
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
44
|
Chen D, Li Y, Wu H, Wu Y, Tang N, Chen S, Liu Y, Wang J, Zhang X, Li Z. Ghrelin-Ghrelin receptor (GSHR) pathway via endocannabinoid signal affects the expression of NPY to promote the food intake of Siberian sturgeon (Acipenser baerii). Horm Behav 2022; 143:105199. [PMID: 35597053 DOI: 10.1016/j.yhbeh.2022.105199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
Abstract
Previous data suggested that activation of endocannabinoid receptor 1 (CB1) was necessary for the orexigenic effect of Ghrelin in rodents, but the information is limited in teleosts. To investigate the feeding regulation pathway of Ghrelin and CB1 in Siberian sturgeon (Acipenser baerii), this study first identified the Ghrelin (345 bp, complete coding sequence) and Ghrelin receptor (GHSR, 500 bp, partial coding sequence) sequences, and then detected their tissue distribution patterns, which showed that Ghrelin is mainly distribution in peripheral tissues, while GSHR is mainly in different brain divisions. Besides, the qPCR before and after feeding showed that the mRNA expressions of Ghrelin and GHSR were inhibited after feeding in telencephalon, diencephalon and mesencephalon. Subsequently, the food intake and appetite factor expressions were measured by i.c.v. co-injection of Ghrelin and GSHR antagonist. The results showed that Ghrelin promoted the food intake of Siberian sturgeon, which was reversed by its receptor antagonist. Besides, i.c.v. injection of Ghrelin decreased telencephalon CART expression while increased NPY expression in the three brain regions. In addition, to further explore the relationship of Ghrelin and CB1 signal regulating feeding, the co-injection of Ghrelin and CB1 antagonists was performed. The results showed that AM6545 (CB1 peripheral restricted antagonist) failed to affect the orexigenic effect of Ghrelin and the expression pattern of NPY mRNA in the telencephalon. While in the diencephalon, the increase of food intake and NPY mRNA expression induced by Ghrelin was completely reversed by Rimonabant (CB1 global antagonist). These results indicate Ghrelin-GSHR pathway promotes the food intake of Siberian sturgeon by inducing the expression of NPY in the diencephalon, and the stimulating effect will be reversed by cannabinoid receptor antagonism. This study provides a foundation for understanding the pathways Ghrelin and CB1 signals in appetite regulation of the teleost.
Collapse
Affiliation(s)
- Defang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China
| | - Yingzi Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China
| | - Hongwei Wu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China; Chengdu Agricultural College, 392#, Detong Bridge Road, Chengdu, China
| | - Yuanbing Wu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China
| | - Ni Tang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China
| | - Shuhuang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China
| | - Youlian Liu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China
| | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, 1124#, Dongtong Road, Neijiang, China
| | - Xin Zhang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China.
| | - Zhiqiong Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, 211#, Huimin Road, Chengdu, China.
| |
Collapse
|
45
|
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: 10] [Impact Index Per Article: 3.3] [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
|
46
|
Staricoff EO, Evans ML. Recent advances in understanding hypothalamic control of defensive responses to hypoglycaemia. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 24:100353. [PMID: 39183767 PMCID: PMC11339540 DOI: 10.1016/j.coemr.2022.100353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Maintenance of normal blood glucose is important for survival. In particular, brain function is dependent on circulating glucose. In health, a series of powerful counterregulatory defences operate to prevent/limit hypoglycaemia. These defences are altered to varying degrees in diabetes and in particular, a subset of people with diabetes can develop profound deficits in these defences placing them at increased risk of suffering episodes of severe hypoglycaemia. Brain is an important controller of glucose homeostasis and developments in molecular techniques have allowed the neurocircuitry of a number of important centrally-controlled homeostatic processes such as energy balance, thirst and thermoregulation to be defined. This review describes how some of these advances have allowed a better understanding of the neuronal/brain ensembles which help protect against hypoglycaemia.
Collapse
Affiliation(s)
- Emily O. Staricoff
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, UK
| | - Mark L. Evans
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, UK
- Department of Medicine, Addenbrookes NIHR Biomedical Campus, Cambridge, UK
| |
Collapse
|
47
|
Cotero V, Graf J, Miwa H, Hirschstein Z, Qanud K, Huerta TS, Tai N, Ding Y, Jimenez-Cowell K, Tomaio JN, Song W, Devarajan A, Tsaava T, Madhavan R, Wallace K, Loghin E, Morton C, Fan Y, Kao TJ, Akhtar K, Damaraju M, Barenboim L, Maietta T, Ashe J, Tracey KJ, Coleman TR, Di Carlo D, Shin D, Zanos S, Chavan SS, Herzog RI, Puleo C. Stimulation of the hepatoportal nerve plexus with focused ultrasound restores glucose homoeostasis in diabetic mice, rats and swine. Nat Biomed Eng 2022; 6:683-705. [PMID: 35361935 PMCID: PMC10127248 DOI: 10.1038/s41551-022-00870-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
Peripheral neurons that sense glucose relay signals of glucose availability to integrative clusters of neurons in the brain. However, the roles of such signalling pathways in the maintenance of glucose homoeostasis and their contribution to disease are unknown. Here we show that the selective activation of the nerve plexus of the hepatic portal system via peripheral focused ultrasound stimulation (pFUS) improves glucose homoeostasis in mice and rats with insulin-resistant diabetes and in swine subject to hyperinsulinemic-euglycaemic clamps. pFUS modulated the activity of sensory projections to the hypothalamus, altered the concentrations of metabolism-regulating neurotransmitters, and enhanced glucose tolerance and utilization in the three species, whereas physical transection or chemical blocking of the liver-brain nerve pathway abolished the effect of pFUS on glucose tolerance. Longitudinal multi-omic profiling of metabolic tissues from the treated animals confirmed pFUS-induced modifications of key metabolic functions in liver, pancreas, muscle, adipose, kidney and intestinal tissues. Non-invasive ultrasound activation of afferent autonomic nerves may represent a non-pharmacologic therapy for the restoration of glucose homoeostasis in type-2 diabetes and other metabolic diseases.
Collapse
Affiliation(s)
- Victoria Cotero
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - John Graf
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Hiromi Miwa
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Khaled Qanud
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tomás S Huerta
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Yuyan Ding
- Yale School of Medicine, New Haven, CT, USA
| | - Kevin Jimenez-Cowell
- Yale School of Medicine, New Haven, CT, USA
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Weiguo Song
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Alex Devarajan
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tea Tsaava
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Radhika Madhavan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Kirk Wallace
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Evelina Loghin
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Christine Morton
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Ying Fan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Tzu-Jen Kao
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | | | | | | | | | - Jeffrey Ashe
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Dino Di Carlo
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Stavros Zanos
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | | | - Chris Puleo
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA.
| |
Collapse
|
48
|
Jais A, Brüning JC. Arcuate Nucleus-Dependent Regulation of Metabolism-Pathways to Obesity and Diabetes Mellitus. Endocr Rev 2022; 43:314-328. [PMID: 34490882 PMCID: PMC8905335 DOI: 10.1210/endrev/bnab025] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 01/12/2023]
Abstract
The central nervous system (CNS) receives information from afferent neurons, circulating hormones, and absorbed nutrients and integrates this information to orchestrate the actions of the neuroendocrine and autonomic nervous systems in maintaining systemic metabolic homeostasis. Particularly the arcuate nucleus of the hypothalamus (ARC) is of pivotal importance for primary sensing of adiposity signals, such as leptin and insulin, and circulating nutrients, such as glucose. Importantly, energy state-sensing neurons in the ARC not only regulate feeding but at the same time control multiple physiological functions, such as glucose homeostasis, blood pressure, and innate immune responses. These findings have defined them as master regulators, which adapt integrative physiology to the energy state of the organism. The disruption of this fine-tuned control leads to an imbalance between energy intake and expenditure as well as deregulation of peripheral metabolism. Improving our understanding of the cellular, molecular, and functional basis of this regulatory principle in the CNS could set the stage for developing novel therapeutic strategies for the treatment of obesity and metabolic syndrome. In this review, we summarize novel insights with a particular emphasis on ARC neurocircuitries regulating food intake and glucose homeostasis and sensing factors that inform the brain of the organismal energy status.
Collapse
Affiliation(s)
- Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, 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, Leipzig, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,National Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
49
|
Tear film and ocular surface neuropeptides: Characteristics, synthesis, signaling and implications for ocular surface and systemic diseases. Exp Eye Res 2022; 218:108973. [PMID: 35149082 DOI: 10.1016/j.exer.2022.108973] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/14/2021] [Accepted: 02/01/2022] [Indexed: 01/13/2023]
Abstract
Ocular surface neuropeptides are vital molecules primarily involved in maintaining ocular surface integrity and homeostasis. They also serve as communication channels between the nervous system and the immune system, maintaining the homeostasis of the ocular surface. Tear film and ocular surface neuropeptides have a role in disease often due to abnormalities in their synthesis (either high or low production), signaling through defective receptors, or both. This creates imbalances in otherwise normal physiological processes. They have been observed to be altered in many ocular surface and systemic diseases including dry eye disease, ocular allergy, keratoconus, LASIK-induced dry eye, pterygium, neurotrophic keratitis, corneal graft rejection, microbial keratitis, headaches and diabetes. This review examines the characteristics of neuropeptides, their synthesis and their signaling through G-protein coupled receptors. The review also explores the types of neuropeptides within the tears and ocular surface, and how they change in ocular and systemic diseases.
Collapse
|
50
|
Qi Y, Lee NJ, Ip CK, Enriquez R, Tasan R, Zhang L, Herzog H. NPY derived from AGRP neurons controls feeding via Y1 and energy expenditure and food foraging behaviour via Y2 signalling. Mol Metab 2022; 59:101455. [PMID: 35167990 PMCID: PMC8886056 DOI: 10.1016/j.molmet.2022.101455] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Aguti-related protein (AGRP) neurons in the arcuate nucleus of the hypothalamus (ARC), which co-express neuropeptide Y (NPY), are key regulators of feeding and energy homeostasis. However, the precise role NPY has within these neurons and the specific pathways that it control are still unclear. In this article, we aimed to determine what aspects of feeding behaviour and energy homeostasis are controlled by NPY originating from AGRP neurons and which Y-receptor pathways are utilised to fulfil this function. Methods Novel conditional Agrpcre/+;Npylox/lox knockout mice were generated and comprehensively phenotyped, both under standard chow as well as high-fat-diet conditions. Designer receptor exclusively activated by designer drugs (DREADD) technology was used to assess the altered responses on feeding and energy homeostasis control in the absence of NPY in these neurons. Rescue experiments utilising Npy1r- and Npy2r-selective NPY ligands were performed to assess which component of the energy homeostasis control is dependent by which specific Y-receptor pathway. Results We show that the specific deletion of Npy only in AGRP neurons leads to a paradoxical mild obese phenotype associated with reduced locomotion and energy expenditure and increased feeding and Respiratory Quotient (RQ) that remain elevated under a positive energy balance. The activation of Npy-deficient AGRP neurons via DREADD's is still able to drive feeding, yet with a delayed onset. Additionally, Clozapine-N-oxide (CNO) treatment reduces locomotion without impacting on energy expenditure. Rescue experiments re-introducing Npy1r- and Npy2r-selective NPY ligands revealed that the increased feeding and RQ are mostly driven by Npy1r, whereas energy expenditure and locomotion are controlled by Npy2r signalling. Conclusion Together, these results demonstrate that NPY originating from AGRP neurons is not only critical to initiate but also for continuously driving feeding, and we for the first time identify which Y-receptor controls which pathway. Neuropeptide Y (NPY) in AGRP neurons is required to initiate and maintain a feeding drive. NPY controls feeding and RQ via Npy1r signalling. NPY controls energy expenditure and activity via Npy2r signalling. Reduced NPY levels in AGRP neurons result in increased Npy2r expression in POMC neurons.
Collapse
|