1
|
Na ES. Epigenetic Mechanisms of Obesity: Insights from Transgenic Animal Models. Life (Basel) 2025; 15:653. [PMID: 40283207 PMCID: PMC12028693 DOI: 10.3390/life15040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Obesity is a chronic disease with prevalence rates that have risen dramatically over the past four decades. This increase is not due to changes in the human genome but rather to environmental factors that promote maladaptive physiological responses. Emerging evidence suggests that external influences, such as high-fat diets, modify the epigenome-the interface between genes and the environment-leading to persistent alterations in energy homeostasis. This review explores the role of epigenetic mechanisms in obesity, emphasizing insights from transgenic animal models and clinical studies. Additionally, we discuss the evolution of obesity research from homeostatic to allostatic frameworks, highlighting key neuroendocrine regulators of energy balance.
Collapse
Affiliation(s)
- Elisa S Na
- School of Social Work, Psychology, & Philosophy, Texas Woman's University, Denton, TX 76209, USA
| |
Collapse
|
2
|
Talbi R, Stincic TL, Ferrari K, Hae CJ, Walec K, Medve E, Gerutshang A, León S, McCarthy EA, Rønnekleiv OK, Kelly MJ, Navarro VM. POMC neurons control fertility through differential signaling of MC4R in Kisspeptin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.18.580873. [PMID: 38915534 PMCID: PMC11195098 DOI: 10.1101/2024.02.18.580873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Inactivating mutations in the melanocortin 4 receptor (MC4R) gene cause monogenic obesity. Interestingly, female patients also display various degrees of reproductive disorders, in line with the subfertile phenotype of MC4RKO female mice. However, the cellular mechanisms by which MC4R regulates reproduction are unknown. Kiss1 neurons directly stimulate gonadotropin-releasing hormone (GnRH) release through two distinct populations; the Kiss1ARH neurons, controlling GnRH pulses, and the sexually dimorphic Kiss1AVPV/PeN neurons controlling the preovulatory LH surge. Here, we show that Mc4r expressed in Kiss1 neurons regulates fertility in females. In vivo, deletion of Mc4r from Kiss1 neurons in female mice replicates the reproductive impairments of MC4RKO mice without inducing obesity. Conversely, reinsertion of Mc4r in Kiss1 neurons of MC4R null mice restores estrous cyclicity and LH pulsatility without reducing their obese phenotype. In vitro, we dissect the specific action of MC4R on Kiss1ARH vs Kiss1AVPV/PeN neurons and show that MC4R activation excites Kiss1ARH neurons through direct synaptic actions. In contrast, Kiss1AVPV/PeN neurons are normally inhibited by MC4R activation except under elevated estradiol levels, thus facilitating the activation of Kiss1AVPV/PeN neurons to induce the LH surge driving ovulation in females. Our findings demonstrate that POMCARH neurons acting through MC4R, directly regulate reproductive function in females by stimulating the "pulse generator" activity of Kiss1ARH neurons and restricting the activation of Kiss1AVPV/PeN neurons to the time of the estradiol-dependent LH surge, and thus unveil a novel pathway of the metabolic regulation of fertility by the melanocortin system.
Collapse
Affiliation(s)
- Rajae Talbi
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Todd L. Stincic
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Kaitlin Ferrari
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Choi Ji Hae
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Karol Walec
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth Medve
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Achi Gerutshang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Silvia León
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth A. McCarthy
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Oline K. Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Martin J. Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Víctor M. Navarro
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Program in Neuroscience, Boston, MA, USA
| |
Collapse
|
3
|
Yu H, Feng N, Zhong W, Han Y, Cheng Y, Zhang Z, Wang Y, Gao P, Huang R, Zhang C, Liu Z, Dong J, He Z, Lai H, Shen Z, Zhai Q. Nmnat2 deficiency in the arcuate nucleus or paraventricular nucleus induces Sarm1-independent neuron loss and liraglutide-reversible obesity. FASEB J 2025; 39:e70400. [PMID: 39964232 DOI: 10.1096/fj.202402546r] [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: 10/22/2024] [Revised: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 05/10/2025]
Abstract
Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) plays an important role in maintaining axon integrity, and the arcuate nucleus (ARC), and paraventricular nucleus (PVN) are crucial nuclei in the control of energy balance. However, the effect of Nmnat2 deficiency in ARC and PVN is still unclear. Nmnat2 loxP/loxP or Nmnat2 loxP/loxP , Sarm1 -/- mice were bilaterally injected with AAV-CMV-GFP-Cre once into the ARC, PVN, or lateral parabrachial nucleus (LPBN) to obtain Nmnat2 ARC-/- , Nmnat2 PVN-/- , Nmnat2 LPBN-/- , Nmnat2 ARC-/- , SKO, Nmnat2 PVN-/- , SKO, or Nmnat2 LPBN-/- , SKO mice. Syn1-Cre mice were bilaterally injected with AAV-EF1a-flex-taCasp3-TEVp once into the ARC or PVN to specifically induce neuron loss. Metabolic changes were measured in the mice intraperitoneally injected with or without liraglutide, a glucagon-like peptide-1 (GLP-1) analog. Neuron loss and neuron activation were monitored by immunofluorescence. Deletion of Nmnat2 in ARC or PVN of mice leads to neuron loss, increased food intake, and obesity in a Sarm1-independent manner. Intraperitoneal injection of liraglutide activates neurons in PVN and LPBN, and attenuates hyperphagia and obesity induced by Nmnat2 deletion or apoptosis of Syn1-positive neurons in ARC or PVN, but has no significant effect on neuron loss. Nmnat2 deficiency in LPBN leads to death within 2 weeks, which can be markedly rescued by Sarm1 deficiency. These data show that deletion of Nmnat2 in ARC or PVN in adult mice leads to Sarm1-independent neuron loss, and liraglutide-reversible hyperphagia and obesity. These findings also elucidate the integrated role of ARC or PVN for downregulating food intake, the requirement of LPBN for survival, and the ARC- or PVN-independent effect of GLP-1 on food intake.
Collapse
Affiliation(s)
- Huimin Yu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ning Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wuling Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yumo Han
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yalan Cheng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhentong Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingqi Wang
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peidong Gao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cong Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zongyang Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jieya Dong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhishui He
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hejin Lai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ziru Shen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
4
|
Haddad HK, Mercado-Reyes JI, Mustafá ER, D’Souza SP, Chung CS, Nestor RRM, Olinski LE, Martinez Damonte V, Saskin J, Vemaraju S, Raingo J, Kauer JA, Lang RA, Oancea E. Hypothalamic opsin 3 suppresses MC4R signaling and potentiates Kir7.1 to promote food consumption. Proc Natl Acad Sci U S A 2025; 122:e2403891122. [PMID: 39951488 PMCID: PMC11874419 DOI: 10.1073/pnas.2403891122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 12/02/2024] [Indexed: 02/16/2025] Open
Abstract
Mammalian opsin 3 (OPN3) is a member of the opsin family of G-protein-coupled receptors with ambiguous light sensitivity. OPN3 was first identified in the brain (and named encephalopsin) and subsequently found to be expressed in other tissues. In adipocytes, OPN3 is necessary for light responses that modulate lipolysis and glucose uptake, while OPN3 in human skin melanocytes regulates pigmentation in a light-independent manner. Despite its initial discovery in the brain, OPN3 functional mechanisms in the brain remain elusive. Here, we investigated the molecular mechanism of OPN3 function in the paraventricular nucleus (PVN) of the hypothalamus. We show that Opn3 is coexpressed with the melanocortin 4 receptor (Mc4r) in a population of PVN neurons, where it negatively regulates MC4R-mediated cAMP signaling in a specific and Gαi/o-dependent manner. Under baseline conditions, OPN3 via Gαi/o potentiates the activity of the inward rectifying Kir7.1 channel, previously shown to be closed in response to agonist-mediated activation of MC4R in a Gαs-independent manner. In mice, we found that Opn3 in Mc4r-expressing neurons regulates food consumption. Our results reveal the first mechanistic insight into OPN3 function in the hypothalamus, uncovering a unique mechanism by which OPN3 functions to potentiate Kir7.1 activity and negatively regulate MC4R-mediated cAMP signaling, thereby promoting food intake.
Collapse
Affiliation(s)
- Hala K. Haddad
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Jonathan I. Mercado-Reyes
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - E. Román Mustafá
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - C. Sean Chung
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Ramses R. M. Nestor
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Lauren E. Olinski
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Joshua Saskin
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Shruti Vemaraju
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Jesica Raingo
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Julie A. Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH45229
| | - Elena Oancea
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| |
Collapse
|
5
|
Qiu T, Fu O. GPCRs in hypothalamic neurons and their roles in controlling food intake and metabolism. Front Mol Neurosci 2025; 18:1536577. [PMID: 39974186 PMCID: PMC11835808 DOI: 10.3389/fnmol.2025.1536577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
G-protein coupled receptor (GPCR) subtypes within the hypothalamus play a pivotal role in maintaining body homeostasis, particularly in the regulation of food intake and energy metabolism. This review provides an overview of classical loss and gain-of-function studies on GPCRs related to feeding and metabolism, with a focus on emerging cell-type-specific investigations. These studies reveal that diverse GPCR-expressing neuronal populations are intricately linked to feeding and energy balance. We also discuss recent findings that highlight the interaction of distinct peptide-GPCR systems in modulating complex feeding behaviors.
Collapse
Affiliation(s)
- Tian Qiu
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- Laboratory of Food Perception Science, Science Center for Future Foods, Jiangnan University, Wuxi, Jiangsu, China
| | - Ou Fu
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- Laboratory of Food Perception Science, Science Center for Future Foods, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
Zhang M, Wang Q, Wang Y. Brain endocannabinoid control of metabolic and non-metabolic feeding behaviors. Neurochem Int 2025; 183:105921. [PMID: 39708909 DOI: 10.1016/j.neuint.2024.105921] [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: 10/14/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The central endocannabinoid (eCB) system in brain shows a crucial role in the regulation of feeding behaviors, influencing both metabolic and non-metabolic mechanisms of appetite control, which has been paid much attention. Although there are already many review articles discussing eCB modulation of feeding behaviors, our paper attempts to summarize the recent advancements through synapses, circuits, and network in brain. Our focus is on the dual role of eCB signalling in regulating metabolic energy balance and hedonic reward-related feeding. In the context of metabolic regulation of feeding behaviors, eCBs affect the hypothalamic circuits that balance hunger and satiety through signal integration related to energy status and nutrient availability. Dysregulation of this system can contribute to metabolic disorders such as obesity and anorexia. In non-metabolic feeding, the eCB system influences the hedonic aspects of eating by modulating reward pathways, including the mesolimbic system and the olfactory bulb, critical for motivating food intake and processing sensory cues. This review also explores therapeutic strategies targeting the eCB system, including cannabinoid receptor antagonists and eCB hydrolase enzyme inhibitors, which hold promise for treating conditions associated with appetite dysregulation and eating disorders. By synthesizing recent findings, we aim to highlight the intricate mechanisms through which the eCB system affects feeding behavior and to propose future directions for research and therapeutic intervention in the realm of appetite control and eating disorders.
Collapse
Affiliation(s)
- Maoxing Zhang
- Basic School of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 260071, China
| | - Qingyu Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ying Wang
- Basic School of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 260071, China.
| |
Collapse
|
7
|
Zhang QJ, Luan JC, Gu Q, Song NH, Xia JD. Leptin action on ARC-PVN neural circuit regulates ejaculation behavior by altering sympathetic neuroplasticity. Andrology 2025. [PMID: 39748719 DOI: 10.1111/andr.13833] [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: 06/02/2024] [Revised: 09/08/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Although some studies have revealed the close relationship between leptin and premature ejaculation in clinical practice, whether and how leptin participates in the regulation of ejaculatory behaviors are still unknown. OBJECTIVE To explore the role of leptin on ejaculatory behaviors and its underlying mechanism. MATERIALS AND METHODS Copulation behavior tests were performed after acute and chronic leptin administration at peripheral and central levels. To compare changes in sympathetic nervous system activity, lumbar sympathetic nervous activity, serum noradrenaline levels, and the distribution of sympathetic fibers in vas deferens and seminal vesicles were analyzed. Construction of virus vector, immunohistochemistry, and optogenetics techniques were used to explore the neural circuit mechanism. The density of dendritic spines in parvocellular region of paraventricular nucleus was measured by Golgi staining. RESULTS Acute administration of leptin had no effect on ejaculation behavior in male mice. However, both mount latency and ejaculation latency were significantly shortened, even if serum leptin decreased to normal level, after chronic administration of leptin at peripheral or central level. Additionally, sympathetic fibers in vas deferens and seminal vesicles obviously increased, in which arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus played an important role. Dendritic spine density in parvocellular region increased after chronic leptin administration. DISCUSSION AND CONCLUSION The role of leptin in regulating ejaculation behavior was chronic, not acute, in which leptin chronically modulated sympathetic neuroplasticity via arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus and promoted ejaculatory behaviors. Increased dendritic spine density in parvocellular region of paraventricular nucleus may be involved as well.
Collapse
Affiliation(s)
- Qi-Jie Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Jiao-Chen Luan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Gu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ning-Hong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Dong Xia
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Farooqi S. Understanding the desire for dessert. Science 2025; 387:717-718. [PMID: 39946484 DOI: 10.1126/science.adv4359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
A neural circuit in mice mediates the preference for high-sugar food after a meal.
Collapse
Affiliation(s)
- Sadaf Farooqi
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Sunkavalli PS, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Stochastic neuropeptide signals compete to calibrate the rate of satiation. Nature 2025; 637:137-144. [PMID: 39506113 PMCID: PMC11981016 DOI: 10.1038/s41586-024-08164-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Neuropeptides have important roles in neural plasticity, spiking and behaviour1. Yet, many fundamental questions remain regarding their spatiotemporal transmission, integration and functions in the awake brain. Here we examined how MC4R-expressing neurons in the paraventricular nucleus of the hypothalamus (PVHMC4R) integrate neuropeptide signals to modulate feeding-related fast synaptic transmission and titrate the transition to satiety2-6. We show that hunger-promoting AgRP axons release the neuropeptide NPY to decrease the second messenger cAMP in PVHMC4R neurons, while satiety-promoting POMC axons release the neuropeptide αMSH to increase cAMP. Each release event is all-or-none, stochastic and can impact multiple neurons within an approximately 100-µm-diameter region. After release, NPY and αMSH peptides compete to control cAMP-the amplitude and persistence of NPY signalling is blunted by high αMSH in the fed state, while αMSH signalling is blunted by high NPY in the fasted state. Feeding resolves this competition by simultaneously elevating αMSH release and suppressing NPY release7,8, thereby sustaining elevated cAMP in PVHMC4R neurons throughout a meal. In turn, elevated cAMP facilitates potentiation of feeding-related excitatory inputs with each bite to gradually promote satiation across many minutes. Our findings highlight biochemical modes of peptide signal integration and information accumulation to guide behavioural state transitions.
Collapse
Affiliation(s)
- Stephen X Zhang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Angela Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Paula K Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lauren F Christenson
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter N Kalugin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Praneel S Sunkavalli
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Neuroscience Graduate Group, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Akash Pal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Niu Y, Yu W, Kou X, Wu S, Liu M, Chen C, Ji J, Shao Y, Xue Z. Bioactive compounds regulate appetite through the melanocortin system: a review. Food Funct 2024; 15:11811-11833. [PMID: 39506527 DOI: 10.1039/d4fo04024d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Obesity, a significant health crisis, arises from an imbalance between energy intake and expenditure. Enhancing appetite regulation has garnered substantial attention from researchers as a novel and effective strategy for weight management. The melanocortin system, situated in the hypothalamus, is recognized as a critical node in the regulation of appetite. It integrates long-term and short-term hormone signals from the periphery as well as nutrients, forming a complex network of interacting feedback mechanisms with the gut-brain axis, significantly contributing to the regulation of energy homeostasis. Appetite regulation by bioactive compounds has been a focus of intensive research due to their favorable safety profiles and easy accessibility. These bioactive compounds, derived from a variety of plant and animal sources, modulate the melanocortin system and influence appetite and energy homeostasis through multiple pathways: central nervous system, peripheral hormones, and intestinal microbiota. Here, we review the anatomy, function, and receptors of the melanocortin system, outline the long-term and short-term regulatory hormones that act on the melanocortin system, and discuss the bioactive compounds and their mechanisms of action that exert a regulatory effect on appetite by targeting the melanocortin system. This review contributes to a better understanding of how bioactive compounds regulate appetite via the melanocortin system, thereby providing nutritional references for citizens' dietary preferences.
Collapse
Affiliation(s)
- Yujia Niu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Wancong Yu
- Biotechnology Research Institute, Tianjin Academy of Agricultural Sciences, Tianjin 300384, China
| | - Xiaohong Kou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Shuqi Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Mengyi Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Chenlong Chen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Jiaxin Ji
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Ying Shao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| | - Zhaohui Xue
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China.
| |
Collapse
|
11
|
Kosse C, Ivanov J, Knight Z, Pellegrino K, Friedman J. A subcortical feeding circuit linking an interoceptive node to jaw movement. Nature 2024; 636:151-161. [PMID: 39443799 PMCID: PMC11618074 DOI: 10.1038/s41586-024-08098-1] [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: 10/24/2022] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The brain processes an array of stimuli, enabling the selection of appropriate behavioural responses, but the neural pathways linking interoceptive inputs to outputs for feeding are poorly understood1-3. Here we delineate a subcortical circuit in which brain-derived neurotrophic factor (BDNF)-expressing neurons in the ventromedial hypothalamus (VMH) directly connect interoceptive inputs to motor centres, controlling food consumption and jaw movements. VMHBDNF neuron inhibition increases food intake by gating motor sequences of feeding through projections to premotor areas of the jaw. When food is unavailable, VMHBDNF inhibition elicits consummatory behaviours directed at inanimate objects such as wooden blocks, and inhibition of perimesencephalic trigeminal area (pMe5) projections evokes rhythmic jaw movements. The activity of these neurons is decreased during food consumption and increases when food is in proximity but not consumed. Activity is also increased in obese animals and after leptin treatment. VMHBDNF neurons receive monosynaptic inputs from both agouti-related peptide (AgRP) and proopiomelanocortin neurons in the arcuate nucleus (Arc), and constitutive VMHBDNF activation blocks the orexigenic effect of AgRP activation. These data indicate an Arc → VMHBDNF → pMe5 circuit that senses the energy state of an animal and regulates consummatory behaviours in a state-dependent manner.
Collapse
Affiliation(s)
- Christin Kosse
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Jessica Ivanov
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Zachary Knight
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Pellegrino
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Jeffrey Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
12
|
Bombassaro B, Araujo EP, Velloso LA. The hypothalamus as the central regulator of energy balance and its impact on current and future obesity treatments. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240082. [PMID: 39876968 PMCID: PMC11771753 DOI: 10.20945/2359-4292-2024-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 01/31/2025]
Abstract
The hypothalamus is a master regulator of energy balance in the body. First-order hypothalamic neurons localized in the arcuate nucleus sense systemic signals that indicate the energy stores in the body. Through distinct projections, arcuate nucleus neurons communicate with second-order neurons, which are mostly localized in the paraventricular nucleus and in the lateral hypothalamus. The signals then proceed to third- and fourth-order neurons that activate complex responses aimed at maintaining whole-body energy homeostasis. During the last 30 years, since the identification of leptin in 1994, there has been a great advance in the unveiling of the hypothalamic and extra-hypothalamic neuronal networks that control energy balance. This has contributed to the characterization of the mechanisms by which glucagon-like peptide-1 receptor agonists promote body mass reduction and has opened new windows of opportunity for the development of drugs to treat obesity. This review presents an overview of the mechanisms involved in the hypothalamic regulation of energy balance and discusses how advancements in this field are contributing to the development of new pharmacological strategies to treat obesity.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Eliana P Araujo
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Licio A Velloso
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| |
Collapse
|
13
|
Mahishi D, Agrawal N, Jiang W, Yapici N. From Mammals to Insects: Exploring the Genetic and Neural Basis of Eating Behavior. Annu Rev Genet 2024; 58:455-485. [PMID: 39585905 DOI: 10.1146/annurev-genet-111523-102202] [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] [Indexed: 11/27/2024]
Abstract
Obesity and anorexia are life-threatening diseases that are still poorly understood at the genetic and neuronal levels. Patients suffering from these conditions experience disrupted regulation of food consumption, leading to extreme weight gain or loss and, in severe situations, death from metabolic dysfunction. Despite the development of various behavioral and pharmacological interventions, current treatments often yield limited and short-lived success. To address this, a deeper understanding of the genetic and neural mechanisms underlying food perception and appetite regulation is essential for identifying new drug targets and developing more effective treatment methods. This review summarizes the progress of past research in understanding the genetic and neural mechanisms controlling food consumption and appetite regulation, focusing on two key model organisms: the fruit fly Drosophila melanogaster and the mouse Mus musculus. These studies investigate how the brain senses energy and nutrient deficiency, how sensory signals trigger appetitive behaviors, and how food intake is regulated through interconnected neural circuits in the brain.
Collapse
Affiliation(s)
- Deepthi Mahishi
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Naman Agrawal
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Wenshuai Jiang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| |
Collapse
|
14
|
Xing M, Li Y, Zhang Y, Zhou J, Ma D, Zhang M, Tang M, Ouyang T, Zhang F, Shi X, Sun J, Chen Z, Zhang WJ, Zhang S, Xie X. Paraventricular hypothalamic RUVBL2 neurons suppress appetite by enhancing excitatory synaptic transmission in distinct neurocircuits. Nat Commun 2024; 15:8939. [PMID: 39414808 PMCID: PMC11484884 DOI: 10.1038/s41467-024-53258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The paraventricular hypothalamus (PVH) is crucial for food intake control, yet the presynaptic mechanisms underlying PVH neurons remain unclear. Here, we show that RUVBL2 in the PVH is significantly reduced during energy deficit, and knockout (KO) of PVH RUVBL2 results in hyperphagic obesity in mice. RUVBL2-expressing neurons in the PVH (PVHRUVBL2) exert the anorexigenic effect by projecting to the arcuate hypothalamus, the dorsomedial hypothalamus, and the parabrachial complex. We further demonstrate that PVHRUVBL2 neurons form the synaptic connections with POMC and AgRP neurons in the ARC. PVH RUVBL2 KO impairs the excitatory synaptic transmission by reducing presynaptic boutons and synaptic vesicles near active zone. Finally, RUVBL2 overexpression in the PVH suppresses food intake and protects against diet induced obesity. Together, this study demonstrates an essential role for PVH RUVBL2 in food intake control, and suggests that modulation of synaptic plasticity could be an effective way to curb appetite and obesity.
Collapse
Affiliation(s)
- Mingming Xing
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Juemou Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Danting Ma
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Mengqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Minglei Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ting Ouyang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Fumiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Weiping J Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
15
|
Liu Y, Lin D, Najam SS, Huang S, Song M, Sirakawin C, Zhao C, Jiang H, Konopka W, Herzig S, Vinnikov IA. Functional redundancy between glucocorticoid and mineralocorticoid receptors in mature corticotropin-releasing hormone neurons protects from obesity. Obesity (Silver Spring) 2024; 32:1885-1896. [PMID: 39315404 DOI: 10.1002/oby.24116] [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: 02/20/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE Here, we aimed to investigate the role of glucocorticoid and mineralocorticoid receptors (GRs and MRs, respectively) in the regulation of energy homeostasis. METHODS We used three mouse models with simultaneous deletion of GRs and MRs in either forebrain neurons, the paraventricular nucleus, or corticotropin-releasing hormone (CRH) neurons and compared them with wild-type controls or isolated knockout groups. In addition to body weight, food intake, energy expenditure, insulin sensitivity, fat/lean mass distribution, and plasma corticosterone levels, we also performed transcriptomic analysis of CRH neurons and assessed their response to melanocortinergic stimulation. RESULTS Similar to global double-knockout models, deletion of GRs and MRs specifically in mature CRH neurons resulted in obesity. Importantly, the latter was accompanied by insulin resistance, but not increased plasma corticosterone levels. Transcriptomic analysis of these neurons revealed upregulation of several genes involved in postsynaptic signal transduction, including the Ptk2b gene, which encodes proline-rich tyrosine kinase 2. Knockout of both nuclear receptors leads to upregulation of Ptk2b in CRH neurons, which results in their diminished responsiveness to melanocortinergic stimulation. CONCLUSIONS Our data demonstrate the functional redundancy of GRs and MRs in CRH neurons to maintain energy homeostasis and prevent obesity. Simultaneous targeting of both receptors might represent an unprecedented approach to counteract obesity.
Collapse
Affiliation(s)
- Yu Liu
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dongfa Lin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Syeda Sadia Najam
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Shangyuan Huang
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Muyi Song
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chaweewan Sirakawin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Catherine Zhao
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Haixia Jiang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Witold Konopka
- Laboratory of Neuroplasticity and Metabolism, Department of Life Sciences and Biotechnology, Łukasiewicz PORT Polish Center for Technology Development, Wrocław, Poland
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Munich Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany; Molecular Metabolic Control, Technical University Munich, Munich, Germany; German Center for Diabetes Research, Munich, Germany
| | - Ilya A Vinnikov
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Wu Z, Hernandez A. Thyroid Hormone Clearance in the Paraventricular Nucleus of Male Mice Regulates Lean Mass and Physical Activity. Neuroendocrinology 2024; 114:1066-1076. [PMID: 39293416 PMCID: PMC11560500 DOI: 10.1159/000541525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/17/2024] [Indexed: 09/20/2024]
Abstract
INTRODUCTION The actions of thyroid hormones (THs) in the central nervous system are relevant to food intake and energy expenditure. TH receptors exhibit high expression in brain areas modulating energy balance, including the arcuate, paraventricular (PVN), supraoptic, and ventromedial (VMH) hypothalamic nuclei. METHODS To examine the role of THs in the regulation of energy balance via action in specific hypothalamic nuclei of the adult mouse, we performed experiments of conditional inactivation of DIO3, the enzyme responsible for the clearance of THs, in the lateral hypothalamus (LH), and VMH and PVN hypothalamic nuclei. We accomplished DIO3 genetic inactivation via stereotaxic injection of the AAV-cre vector into adult mice homozygous for a "floxed" Dio3 allele. RESULTS Dio3 inactivation in the LH and VMH of males or females did not result in significant changes in body weight 8 weeks after injection. However, inactivation of Dio3 in the PVN resulted in increased body weight (both fat mass and lean mass) and locomotor activity, and decreased hypothalamic Mc4r expression in male, but not female mice. However, PNV-specific Dio3 KO did not cause hyperphagia. CONCLUSION These results suggest local TH action influences MC4R signaling and possibly other PVN-associated circuitries, with consequences for body composition and energy balance endpoints, but not for orexigenic pathways. They also support a regulatory role for PVN Dio3 in the central regulation of energy homeostasis in adult life.
Collapse
Affiliation(s)
- Zhaofei Wu
- Department of Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074
| | - Arturo Hernandez
- Department of Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074
| |
Collapse
|
17
|
Li Y, Kim M, Jiang L, Baron L, Faulkner LD, Olson DP, Li X, Gannot N, Li P, Rui L. SH2B1 Defends Against Energy Imbalance, Obesity, and Metabolic Disease via a Paraventricular Hypothalamus→Dorsal Raphe Nucleus Neurocircuit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400437. [PMID: 38885417 PMCID: PMC11336965 DOI: 10.1002/advs.202400437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/01/2024] [Indexed: 06/20/2024]
Abstract
SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Min‐Hyun Kim
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- College of Health SolutionsArizona State UniversityPhoenixAZ85004USA
| | - Lin Jiang
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Lorelei Baron
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Latrice D. Faulkner
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - David P. Olson
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Xingyu Li
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Noam Gannot
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Peng Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Liangyou Rui
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| |
Collapse
|
18
|
Griffin H, Hanson J, Phelan KD, Baldini G. MC4R Localizes at Excitatory Postsynaptic and Peri-Postsynaptic Sites of Hypothalamic Neurons in Primary Culture. Cells 2024; 13:1235. [PMID: 39120267 PMCID: PMC11311852 DOI: 10.3390/cells13151235] [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: 04/28/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain locations encompassing the hypothalamus and the brainstem, where the receptor controls several body functions, including metabolism. In a well-defined pathway to decrease appetite, hypothalamic proopiomelanocortin (POMC) neurons localized in the arcuate nucleus (Arc) project to MC4R neurons in the paraventricular nuclei (PVN) to release the natural MC4R agonist α-melanocyte-stimulating hormone (α-MSH). Arc neurons also project excitatory glutamatergic fibers to the MC4R neurons in the PVN for a fast synaptic transmission to regulate a satiety pathway potentiated by α-MSH. By using super-resolution microscopy, we found that in hypothalamic neurons in a primary culture, postsynaptic density protein 95 (PSD95) colocalizes with GluN1, a subunit of the ionotropic N-methyl-D-aspartate receptor (NMDAR). Thus, hypothalamic neurons form excitatory postsynaptic specializations. To study the MC4R distribution at these sites, tagged HA-MC4R under the synapsin promoter was expressed in neurons by adeno-associated virus (AAV) gene transduction. HA-MC4R immunofluorescence peaked at the center and in proximity to the PSD95- and NMDAR-expressing sites. These data provide morphological evidence that MC4R localizes together with glutamate receptors at postsynaptic and peri-postsynaptic sites.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Jude Hanson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| |
Collapse
|
19
|
Van Drunen R, Dai Y, Wei H, Fekry B, Noori S, Shivshankar S, Bravo R, Zhao Z, Yoo SH, Justice N, Wu JQ, Tong Q, Eckel-Mahan K. Cell-specific regulation of the circadian clock by BMAL1 in the paraventricular nucleus: Implications for regulation of systemic biological rhythms. Cell Rep 2024; 43:114380. [PMID: 38935503 PMCID: PMC11446153 DOI: 10.1016/j.celrep.2024.114380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/28/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Circadian rhythms are internal biological rhythms driving temporal tissue-specific, metabolic programs. Loss of the circadian transcription factor BMAL1 in the paraventricular nucleus (PVN) of the hypothalamus reveals its importance in metabolic rhythms, but its functions in individual PVN cells are poorly understood. Here, loss of BMAL1 in the PVN results in arrhythmicity of processes controlling energy balance and alters peripheral diurnal gene expression. BMAL1 chromatin immunoprecipitation sequencing (ChIP-seq) and single-nucleus RNA sequencing (snRNA-seq) reveal its temporal regulation of target genes, including oxytocin (OXT), and restoring circulating OXT peaks in BMAL1-PVN knockout (KO) mice rescues absent activity rhythms. While glutamatergic neurons undergo day/night changes in expression of genes involved in cell morphogenesis, astrocytes and oligodendrocytes show gene expression changes in cytoskeletal organization and oxidative phosphorylation. Collectively, our findings show diurnal gene regulation in neuronal and non-neuronal PVN cells and that BMAL1 contributes to diurnal OXT secretion, which is important for systemic diurnal rhythms.
Collapse
Affiliation(s)
- Rachel Van Drunen
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yulin Dai
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haichao Wei
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Baharan Fekry
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sina Noori
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Samay Shivshankar
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Rafael Bravo
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Seung-Hee Yoo
- MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biochemistry and Cell Biology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nicholas Justice
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jia Qian Wu
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qingchun Tong
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kristin Eckel-Mahan
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Mishra D, Richard JE, Maric I, Shevchouk OT, Börchers S, Eerola K, Krieger JP, Skibicka KP. Lateral parabrachial nucleus astrocytes control food intake. Front Endocrinol (Lausanne) 2024; 15:1389589. [PMID: 38887265 PMCID: PMC11180714 DOI: 10.3389/fendo.2024.1389589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/03/2024] [Indexed: 06/20/2024] Open
Abstract
Food intake behavior is under the tight control of the central nervous system. Most studies to date focus on the contribution of neurons to this behavior. However, although previously overlooked, astrocytes have recently been implicated to play a key role in feeding control. Most of the recent literature has focused on astrocytic contribution in the hypothalamus or the dorsal vagal complex. The contribution of astrocytes located in the lateral parabrachial nucleus (lPBN) to feeding behavior control remains poorly understood. Thus, here, we first investigated whether activation of lPBN astrocytes affects feeding behavior in male and female rats using chemogenetic activation. Astrocytic activation in the lPBN led to profound anorexia in both sexes, under both ad-libitum feeding schedule and after a fasting challenge. Astrocytes have a key contribution to glutamate homeostasis and can themselves release glutamate. Moreover, lPBN glutamate signaling is a key contributor to potent anorexia, which can be induced by lPBN activation. Thus, here, we determined whether glutamate signaling is necessary for lPBN astrocyte activation-induced anorexia, and found that pharmacological N-methyl D-aspartate (NMDA) receptor blockade attenuated the food intake reduction resulting from lPBN astrocyte activation. Since astrocytes have been shown to contribute to feeding control by modulating the feeding effect of peripheral feeding signals, we further investigated whether lPBN astrocyte activation is capable of modulating the anorexic effect of the gut/brain hormone, glucagon like peptide -1, as well as the orexigenic effect of the stomach hormone - ghrelin, and found that the feeding effect of both signals is modulated by lPBN astrocytic activation. Lastly, we found that lPBN astrocyte activation-induced anorexia is affected by a diet-induced obesity challenge, in a sex-divergent manner. Collectively, current findings uncover a novel role for lPBN astrocytes in feeding behavior control.
Collapse
Affiliation(s)
- Devesh Mishra
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jennifer E. Richard
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Ivana Maric
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Olesya T. Shevchouk
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Stina Börchers
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Kim Eerola
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jean-Philippe Krieger
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich - VetSuisse, Zurich, Switzerland
| | - Karolina P. Skibicka
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
21
|
Liu H, Liu Z, Wong HK, Xu N, Liu Q, Li Y, Liu Y, Wong H, Burt ME, Jossy SV, Han J, He Y. Therapeutic Strategies Against Metabolic Imbalance in a Male Mouse Model With 5-HT2CR Loss-of-Function. Endocrinology 2024; 165:bqae063. [PMID: 38815086 DOI: 10.1210/endocr/bqae063] [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: 04/08/2024] [Revised: 05/07/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
The serotonin 2C receptor (5-HT2CR)-melanocortin pathway plays well-established roles in the regulation of feeding behavior and body weight homeostasis. Dysfunctions in this system, such as loss-of-function mutations in the Htr2c gene, can lead to hyperphagia and obesity. In this study, we aimed to investigate the potential therapeutic strategies for ameliorating hyperphagia, hyperglycemia, and obesity associated with a loss-of-function mutation in the Htr2c gene (Htr2cF327L/Y). We demonstrated that reexpressing functional 5-HT2CR solely in hypothalamic pro-opiomelanocortin (POMC) neurons is sufficient to reduce food intake and body weight in Htr2cF327L/Y mice subjected to a high-fat diet (HFD). In addition, 5-HT2CR expression restores the responsiveness of POMC neurons to lorcaserin, a selective agonist for 5-HT2CR. Similarly, administration of melanotan II, an agonist of the melanocortin receptor 4 (MC4R), effectively suppresses feeding and weight gain in Htr2cF327L/Y mice. Strikingly, promoting wheel-running activity in Htr2cF327L/Y mice results in a decrease in HFD consumption and improved glucose homeostasis. Together, our findings underscore the crucial role of the melanocortin system in alleviating hyperphagia and obesity related to dysfunctions of the 5-HT2CR, and further suggest that MC4R agonists and lifestyle interventions might hold promise in counteracting hyperphagia, hyperglycemia, and obesity in individuals carrying rare variants of the Htr2c gene.
Collapse
MESH Headings
- Animals
- Receptor, Serotonin, 5-HT2C/metabolism
- Receptor, Serotonin, 5-HT2C/genetics
- Male
- Mice
- Hyperphagia/metabolism
- Hyperphagia/genetics
- Pro-Opiomelanocortin/metabolism
- Pro-Opiomelanocortin/genetics
- Diet, High-Fat
- Obesity/metabolism
- Obesity/genetics
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/agonists
- alpha-MSH/pharmacology
- alpha-MSH/analogs & derivatives
- Loss of Function Mutation
- Hypothalamus/metabolism
- Body Weight/drug effects
- Eating/drug effects
- Eating/physiology
- Eating/genetics
- Neurons/metabolism
- Neurons/drug effects
- Disease Models, Animal
- Hyperglycemia/metabolism
- Hyperglycemia/genetics
- Mice, Inbred C57BL
- Benzazepines
- Peptides, Cyclic
Collapse
Affiliation(s)
- Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhaoxun Liu
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - HueyXian Kelly Wong
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nathan Xu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qingzhuo Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yongxiang Li
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yao Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - HueyZhong Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Megan E Burt
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sanika V Jossy
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang He
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
Jurisch-Yaksi N, Wachten D, Gopalakrishnan J. The neuronal cilium - a highly diverse and dynamic organelle involved in sensory detection and neuromodulation. Trends Neurosci 2024; 47:383-394. [PMID: 38580512 DOI: 10.1016/j.tins.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/07/2024]
Abstract
Cilia are fascinating organelles that act as cellular antennae, sensing the cellular environment. Cilia gained significant attention in the late 1990s after their dysfunction was linked to genetic diseases known as ciliopathies. Since then, several breakthrough discoveries have uncovered the mechanisms underlying cilia biogenesis and function. Like most cells in the animal kingdom, neurons also harbor cilia, which are enriched in neuromodulatory receptors. Yet, how neuronal cilia modulate neuronal physiology and animal behavior remains poorly understood. By comparing ciliary biology between the sensory and central nervous systems (CNS), we provide new perspectives on the functions of cilia in brain physiology.
Collapse
Affiliation(s)
- Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Science, Norwegian University of Science and Technology, Erling Skalgssons gate 1, 7491 Trondheim, Norway.
| | - Dagmar Wachten
- Department of Biophysical Imaging, Institute of Innate Immunity, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany; Institute for Human Genetics, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07740 Jena, Germany
| |
Collapse
|
23
|
Han Y, Xia G, Harris L, Liu P, Guan D, Wu Q. Reversal of Obesity by Enhancing Slow-wave Sleep via a Prokineticin Receptor Neural Circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591948. [PMID: 38746393 PMCID: PMC11092673 DOI: 10.1101/2024.04.30.591948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Obese subjects often exhibit hypersomnia accompanied by severe sleep fragmentation, while emerging evidence suggests that poor sleep quality promotes overeating and exacerbates diet-induced obesity (DIO). However, the neural circuit and signaling mechanism underlying the reciprocal control of appetite and sleep is yet not elucidated. Here, we report a neural circuit where prokineticin receptor 2 (PROKR2)-expressing neurons within the parabrachial nucleus (PBN) of the brainstem received direct projections from neuropeptide Y receptor Y2 (NPY2R)-expressing neurons within the lateral preoptic area (LPO) of the hypothalamus. The RNA-Seq results revealed Prokr2 in the PBN is the most regulated GPCR signaling gene that is responsible for comorbidity of obesity and sleep dysfunction. Furthermore, those NPY2R LPO neurons are minimally active during NREM sleep and maximally active during wakefulness and REM sleep. Activation of the NPY2R LPO →PBN circuit or the postsynaptic PROKR2 PBN neurons suppressed feeding of a high-fat diet and abrogated morbid sleep patterns in DIO mice. Further studies showed that genetic ablation of the PROKR2 signaling within PROKR2 PBN neurons alleviated the hyperphagia and weight gain, and restored sleep dysfunction in DIO mice. We further discovered pterostilbene, a plant-derived stilbenoid, is a powerful anti-obesity and sleep-improving agent, robustly suppressed hyperphagia and promoted reconstruction of a healthier sleep architecture, thereby leading to significant weight loss. Collectively, our results unveil a neural mechanism for the reciprocal control of appetite and sleep, through which pterostilbene, along with a class of similarly structured compounds, may be developed as effective therapeutics for tackling obesity and sleep disorders.
Collapse
|
24
|
Harvey T, Rios M. The Role of BDNF and TrkB in the Central Control of Energy and Glucose Balance: An Update. Biomolecules 2024; 14:424. [PMID: 38672441 PMCID: PMC11048226 DOI: 10.3390/biom14040424] [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: 02/20/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
The global rise in obesity and related health issues, such as type 2 diabetes and cardiovascular disease, is alarming. Gaining a deeper insight into the central neural pathways and mechanisms that regulate energy and glucose homeostasis is crucial for developing effective interventions to combat this debilitating condition. A significant body of evidence from studies in humans and rodents indicates that brain-derived neurotrophic factor (BDNF) signaling plays a key role in regulating feeding, energy expenditure, and glycemic control. BDNF is a highly conserved neurotrophin that signals via the tropomyosin-related kinase B (TrkB) receptor to facilitate neuronal survival, differentiation, and synaptic plasticity and function. Recent studies have shed light on the mechanisms through which BDNF influences energy and glucose balance. This review will cover our current understanding of the brain regions, neural circuits, and cellular and molecular mechanisms underlying the metabolic actions of BDNF and TrkB.
Collapse
Affiliation(s)
- Theresa Harvey
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
25
|
Metzger PJ, Zhang A, Carlson BA, Sun H, Cui Z, Li Y, Jahnke MT, Layton DR, Gupta MB, Liu N, Kostenis E, Gavrilova O, Chen M, Weinstein LS. A human obesity-associated MC4R mutation with defective Gq/11α signaling leads to hyperphagia in mice. J Clin Invest 2024; 134:e165418. [PMID: 38175730 PMCID: PMC10869179 DOI: 10.1172/jci165418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
Melanocortin 4 receptor (MC4R) mutations are the most common cause of human monogenic obesity and are associated with hyperphagia and increased linear growth. While MC4R is known to activate Gsα/cAMP signaling, a substantial proportion of obesity-associated MC4R mutations do not affect MC4R/Gsα signaling. To further explore the role of specific MC4R signaling pathways in the regulation of energy balance, we examined the signaling properties of one such mutant, MC4R (F51L), as well as the metabolic consequences of MC4RF51L mutation in mice. The MC4RF51L mutation produced a specific defect in MC4R/Gq/11α signaling and led to obesity, hyperphagia, and increased linear growth in mice. The ability of a melanocortin agonist to acutely inhibit food intake when delivered to the paraventricular nucleus (PVN) was lost in MC4RF51L mice, as well as in WT mice in which a specific Gq/11α inhibitor was delivered to the PVN; this provided evidence that a Gsα-independent signaling pathway, namely Gq/11α, significantly contributes to the actions of MC4R on food intake and linear growth. These results suggest that a biased MC4R agonist that primarily activates Gq/11α may be a potential agent to treat obesity with limited untoward cardiovascular and other side effects.
Collapse
Affiliation(s)
| | | | | | - Hui Sun
- Metabolic Diseases Branch and
| | - Zhenzhong Cui
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | | | | | | | - Naili Liu
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Evi Kostenis
- Molecular, Cellular, and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | | |
Collapse
|
26
|
Ameroso D, Rios M. Synaptic plasticity and the role of astrocytes in central metabolic circuits. WIREs Mech Dis 2024; 16:e1632. [PMID: 37833830 PMCID: PMC10842964 DOI: 10.1002/wsbm.1632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023]
Abstract
Neural circuits in the brain, primarily in the hypothalamus, are paramount to the homeostatic control of feeding and energy utilization. They integrate hunger, satiety, and body adiposity cues from the periphery and mediate the appropriate behavioral and physiological responses to satisfy the energy demands of the animal. Notably, perturbations in central homeostatic circuits have been linked to the etiology of excessive feeding and obesity. Considering the ever-changing energy requirements of the animal and required adaptations, it is not surprising that brain-feeding circuits remain plastic in adulthood and are subject to changes in synaptic strength as a consequence of nutritional status. Indeed, synapse density, probability of presynaptic transmitter release, and postsynaptic responses in hypothalamic energy balance centers are tailored to behavioral and physiological responses required to sustain survival. Mounting evidence supports key roles of astrocytes facilitating some of this plasticity. Here we discuss these synaptic plasticity mechanisms and the emerging roles of astrocytes influencing energy and glucose balance control in health and disease. This article is categorized under: Cancer > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
27
|
Aitken CM, Jaramillo JCM, Davis W, Brennan‐Xie L, McDougall SJ, Lawrence AJ, Ryan PJ. Feeding signals inhibit fluid-satiation signals in the mouse lateral parabrachial nucleus to increase intake of highly palatable, caloric solutions. J Neurochem 2023; 167:648-667. [PMID: 37855271 PMCID: PMC10952698 DOI: 10.1111/jnc.15991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
Chemogenetic activation of oxytocin receptor-expressing neurons in the parabrachial nucleus (OxtrPBN neurons) acts as a satiation signal for water. In this research, we investigated the effect of activating OxtrPBN neurons on satiation for different types of fluids. Chemogenetic activation of OxtrPBN neurons in male and female transgenic OxtrCre mice robustly suppressed the rapid, initial (15-min) intake of several solutions after dehydration: water, sucrose, ethanol and saccharin, but only slightly decreased intake of Ensure®, a highly caloric solution (1 kcal/mL; containing 3.72 g protein, 3.27 g fat, 13.42 g carbohydrates, and 1.01 g dietary fibre per 100 mL). OxtrPBN neuron activation also suppressed cumulative, longer-term (2-h) intake of lower caloric, less palatable solutions, but not highly caloric, palatable solutions. These results suggest that OxtrPBN neurons predominantly control initial fluid-satiation responses after rehydration, but not longer-term intake of highly caloric, palatable solutions. The suppression of fluid intake was not because of anxiogenesis, but because OxtrPBN neuron activation decreased anxiety-like behaviour. To investigate the role of different PBN subdivisions on the intake of different solutions, we examined FOS as a proxy marker of PBN neuron activation. Different PBN subdivisions were activated by different solutions: the dorsolateral PBN similarly by all fluids; the external lateral PBN by caloric but not non-caloric solutions; and the central lateral PBN primarily by highly palatable solutions, suggesting PBN subdivisions regulate different aspects of fluid intake. To explore the possible mechanisms underlying the minimal suppression of Ensure® after OxtrPBN neuron activation, we demonstrated in in vitro slice recordings that the feeding-associated agouti-related peptide (AgRP) inhibited OxtrPBN neuron firing in a concentration-related manner, suggesting possible inhibition by feeding-related neurocircuitry of fluid satiation neurocircuitry. Overall, this research suggests that although palatable beverages like sucrose- and ethanol-containing beverages activate fluid satiation signals encoded by OxtrPBN neurons, these neurons can be inhibited by hunger-related signals (agouti-related peptide, AgRP), which may explain why these fluids are often consumed in excess of what is required for fluid satiation.
Collapse
Affiliation(s)
- Connor M. Aitken
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Janine C. M. Jaramillo
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Warren Davis
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Liam Brennan‐Xie
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Stuart J. McDougall
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Philip J. Ryan
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
28
|
Herb BR, Glover HJ, Bhaduri A, Colantuoni C, Bale TL, Siletti K, Hodge R, Lein E, Kriegstein AR, Doege CA, Ament SA. Single-cell genomics reveals region-specific developmental trajectories underlying neuronal diversity in the human hypothalamus. SCIENCE ADVANCES 2023; 9:eadf6251. [PMID: 37939194 PMCID: PMC10631741 DOI: 10.1126/sciadv.adf6251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
The development and diversity of neuronal subtypes in the human hypothalamus has been insufficiently characterized. To address this, we integrated transcriptomic data from 241,096 cells (126,840 newly generated) in the prenatal and adult human hypothalamus to reveal a temporal trajectory from proliferative stem cell populations to mature hypothalamic cell types. Iterative clustering of the adult neurons identified 108 robust transcriptionally distinct neuronal subtypes representing 10 hypothalamic nuclei. Pseudotime trajectories provided insights into the genes driving formation of these nuclei. Comparisons to single-cell transcriptomic data from the mouse hypothalamus suggested extensive conservation of neuronal subtypes despite certain differences in species-enriched gene expression. The uniqueness of hypothalamic neuronal lineages was examined developmentally by comparing excitatory lineages present in cortex and inhibitory lineages in ganglionic eminence, revealing both distinct and shared drivers of neuronal maturation across the human forebrain. These results provide a comprehensive transcriptomic view of human hypothalamus development through gestation and adulthood at cellular resolution.
Collapse
Affiliation(s)
- Brian R. Herb
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hannah J. Glover
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Carlo Colantuoni
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tracy L. Bale
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Hodge
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Arnold R. Kriegstein
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seth A. Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Gruber T, Lechner F, Murat C, Contreras RE, Sanchez-Quant E, Miok V, Makris K, Le Thuc O, González-García I, García-Clave E, Althammer F, Krabichler Q, DeCamp LM, Jones RG, Lutter D, Williams RH, Pfluger PT, Müller TD, Woods SC, Pospisilik JA, Martinez-Jimenez CP, Tschöp MH, Grinevich V, García-Cáceres C. High-calorie diets uncouple hypothalamic oxytocin neurons from a gut-to-brain satiation pathway via κ-opioid signaling. Cell Rep 2023; 42:113305. [PMID: 37864798 PMCID: PMC10636643 DOI: 10.1016/j.celrep.2023.113305] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/21/2023] [Accepted: 10/04/2023] [Indexed: 10/23/2023] Open
Abstract
Oxytocin-expressing paraventricular hypothalamic neurons (PVNOT neurons) integrate afferent signals from the gut, including cholecystokinin (CCK), to adjust whole-body energy homeostasis. However, the molecular underpinnings by which PVNOT neurons orchestrate gut-to-brain feeding control remain unclear. Here, we show that mice undergoing selective ablation of PVNOT neurons fail to reduce food intake in response to CCK and develop hyperphagic obesity on a chow diet. Notably, exposing wild-type mice to a high-fat/high-sugar (HFHS) diet recapitulates this insensitivity toward CCK, which is linked to diet-induced transcriptional and electrophysiological aberrations specifically in PVNOT neurons. Restoring OT pathways in diet-induced obese (DIO) mice via chemogenetics or polypharmacology sufficiently re-establishes CCK's anorexigenic effects. Last, by single-cell profiling, we identify a specialized PVNOT neuronal subpopulation with increased κ-opioid signaling under an HFHS diet, which restrains their CCK-evoked activation. In sum, we document a (patho)mechanism by which PVNOT signaling uncouples a gut-brain satiation pathway under obesogenic conditions.
Collapse
Affiliation(s)
- Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA.
| | - Franziska Lechner
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Cahuê Murat
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Raian E Contreras
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eva Sanchez-Quant
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, Neuherberg, Germany
| | - Viktorian Miok
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Konstantinos Makris
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ismael González-García
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Elena García-Clave
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | | | - Quirin Krabichler
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Dominik Lutter
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rhiannan H Williams
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute for Neurogenomics, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Paul T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Research Unit NeuroBiology of Diabetes, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Neurobiology of Diabetes, TUM School of Medicine, Technical University Munich, 80333 Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Department of Pharmacology and Experimental Therapy, Institute for Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls Hospitals and Clinics, Tübingen, Germany
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - John Andrew Pospisilik
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA
| | - Celia P Martinez-Jimenez
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA.
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
30
|
Zhu N, LeDuc CA, Fennoy I, Laferrère B, Doege CA, Shen Y, Chung WK, Leibel RL. Rare predicted loss of function alleles in Bassoon (BSN) are associated with obesity. NPJ Genom Med 2023; 8:33. [PMID: 37865656 PMCID: PMC10590409 DOI: 10.1038/s41525-023-00376-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/02/2023] [Indexed: 10/23/2023] Open
Abstract
Bassoon (BSN) is a component of a hetero-dimeric presynaptic cytomatrix protein that orchestrates neurotransmitter release with Piccolo (PCLO) from glutamatergic neurons throughout the brain. Heterozygous missense variants in BSN have previously been associated with neurodegenerative disorders in humans. We performed an exome-wide association analysis of ultra-rare variants in about 140,000 unrelated individuals from the UK Biobank to search for new genes associated with obesity. We found that rare heterozygous predicted loss of function (pLoF) variants in BSN are associated with higher BMI with p-value of 3.6e-12 in the UK biobank cohort. Additionally, we identified two individuals (one of whom has a de novo variant) with a heterozygous pLoF variant in a cohort of early onset or extreme obesity and report the clinical histories of these individuals with non-syndromic obesity with no history of neurobehavioral or cognitive disability. The BMI association was replicated in the All of Us whole genome sequencing data. Heterozygous pLoF BSN variants constitute a new etiology for obesity.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles A LeDuc
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- NY Obesity Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Ilene Fennoy
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Blandine Laferrère
- NY Obesity Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Claudia A Doege
- NY Obesity Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
- JP Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
- NY Obesity Research Center, Columbia University Irving Medical Center, New York, NY, USA.
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
| | - Rudolph L Leibel
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
- NY Obesity Research Center, Columbia University Irving Medical Center, New York, NY, USA.
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
31
|
Rupp AC, Tomlinson AJ, Affinati AH, Yacawych WT, Duensing AM, True C, Lindsley SR, Kirigiti MA, MacKenzie A, Polex-Wolf J, Li C, Knudsen LB, Seeley RJ, Olson DP, Kievit P, Myers MG. Suppression of food intake by Glp1r/Lepr-coexpressing neurons prevents obesity in mouse models. J Clin Invest 2023; 133:e157515. [PMID: 37581939 PMCID: PMC10541203 DOI: 10.1172/jci157515] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 08/03/2023] [Indexed: 08/17/2023] Open
Abstract
The adipose-derived hormone leptin acts via its receptor (LepRb) in the brain to control energy balance. A potentially unidentified population of GABAergic hypothalamic LepRb neurons plays key roles in the restraint of food intake and body weight by leptin. To identify markers for candidate populations of LepRb neurons in an unbiased manner, we performed single-nucleus RNA-Seq of enriched mouse hypothalamic LepRb cells, identifying several previously unrecognized populations of hypothalamic LepRb neurons. Many of these populations displayed strong conservation across species, including GABAergic Glp1r-expressing LepRb (LepRbGlp1r) neurons, which expressed more Lepr than other LepRb cell populations. Ablating Lepr from LepRbGlp1r cells provoked hyperphagic obesity without impairing energy expenditure. Similarly, improvements in energy balance caused by Lepr reactivation in GABA neurons of otherwise Lepr-null mice required Lepr expression in GABAergic Glp1r-expressing neurons. Furthermore, restoration of Glp1r expression in LepRbGlp1r neurons in otherwise Glp1r-null mice enabled food intake suppression by the GLP1R agonist, liraglutide. Thus, the conserved GABAergic LepRbGlp1r neuron population plays crucial roles in the suppression of food intake by leptin and GLP1R agonists.
Collapse
Affiliation(s)
| | | | | | - Warren T. Yacawych
- Department of Internal Medicine and
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison M. Duensing
- Department of Internal Medicine and
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cadence True
- Oregon National Primate Research Center, Beaverton, Oregon, USA
| | | | | | | | | | - Chien Li
- Novo Nordisk, Copenhagen, Denmark
| | | | | | - David P. Olson
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul Kievit
- Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Martin G. Myers
- Department of Internal Medicine and
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
Ross RA, Kim A, Das P, Li Y, Choi YK, Thompson AT, Douglas E, Subramanian S, Ramos K, Callahan K, Bolshakov VY, Ressler KJ. Prefrontal cortex melanocortin 4 receptors (MC4R) mediate food intake behavior in male mice. Physiol Behav 2023; 269:114280. [PMID: 37369302 PMCID: PMC10528493 DOI: 10.1016/j.physbeh.2023.114280] [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: 04/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Melanocortin 4 receptor (MC4R) activity in the hypothalamus is crucial for regulation of metabolism and food intake. The peptide ligands for the MC4R are associated with feeding, energy expenditure, and also with complex behaviors that orchestrate energy intake and expenditure, but the downstream neuroanatomical and neurochemical targets associated with these behaviors are elusive. In addition to strong expression in the hypothalamus, the MC4R is highly expressed in the medial prefrontal cortex, a region involved in executive function and decision-making. METHODS Using viral techniques in genetically modified male mice combined with molecular techniques, we identify and define the effects on feeding behavior of a novel population of MC4R expressing neurons in the infralimbic (IL) region of the cortex. RESULTS Here, we describe a novel population of MC4R-expressing neurons in the IL of the mouse prefrontal cortex that are glutamatergic, receive input from melanocortinergic neurons, and project to multiple regions that coordinate appetitive responses to food-related stimuli. The neurons are stimulated by application of MC4R-specific peptidergic agonist, THIQ. Deletion of MC4R from the IL neurons causes increased food intake and body weight gain and impaired executive function in simple food-related behavior tasks. CONCLUSION Together, these data suggest that MC4R neurons of the IL play a critical role in the regulation of food intake in male mice.
Collapse
Affiliation(s)
- Rachel A Ross
- Departments of Neuroscience and Psychiatry, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Psychiatry, McLean Hospital, Boston, MA, USA.
| | - Angela Kim
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Priyanka Das
- Departments of Neuroscience and Psychiatry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yan Li
- Department of Psychiatry, McLean Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Kat Ramos
- Northeastern University, Boston, MA, USA
| | - Kathryn Callahan
- Departments of Neuroscience and Psychiatry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vadim Y Bolshakov
- Department of Psychiatry, McLean Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Kerry J Ressler
- Department of Psychiatry, McLean Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Cho D, O'Berry K, Possa-Paranhos IC, Butts J, Palanikumar N, Sweeney P. Paraventricular Thalamic MC3R Circuits Link Energy Homeostasis with Anxiety-Related Behavior. J Neurosci 2023; 43:6280-6296. [PMID: 37591737 PMCID: PMC10490510 DOI: 10.1523/jneurosci.0704-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023] Open
Abstract
The hypothalamic melanocortin system is critically involved in sensing stored energy and communicating this information throughout the brain, including to brain regions controlling motivation and emotion. This system consists of first-order agouti-related peptide (AgRP) and pro-opiomelanocortin (POMC) neurons located in the hypothalamic arcuate nucleus and downstream neurons containing the melanocortin-3 (MC3R) and melanocortin-4 receptor (MC4R). Although extensive work has characterized the function of downstream MC4R neurons, the identity and function of MC3R-containing neurons are poorly understood. Here, we used neuroanatomical and circuit manipulation approaches in mice to identify a novel pathway linking hypothalamic melanocortin neurons to melanocortin-3 receptor neurons located in the paraventricular thalamus (PVT) in male and female mice. MC3R neurons in PVT are innervated by hypothalamic AgRP and POMC neurons and are activated by anorexigenic and aversive stimuli. Consistently, chemogenetic activation of PVT MC3R neurons increases anxiety-related behavior and reduces feeding in hungry mice, whereas inhibition of PVT MC3R neurons reduces anxiety-related behavior. These studies position PVT MC3R neurons as important cellular substrates linking energy status with neural circuitry regulating anxiety-related behavior and represent a promising potential target for diseases at the intersection of metabolism and anxiety-related behavior such as anorexia nervosa.SIGNIFICANCE STATEMENT Animals must constantly adapt their behavior to changing internal and external challenges, and impairments in appropriately responding to these challenges are a hallmark of many neuropsychiatric disorders. Here, we demonstrate that paraventricular thalamic neurons containing the melanocortin-3 receptor respond to energy-state-related information and external challenges to regulate anxiety-related behavior in mice. Thus, these neurons represent a potential target for understanding the neurobiology of disorders at the intersection of metabolism and psychiatry such as anorexia nervosa.
Collapse
Affiliation(s)
- Dajin Cho
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Kyle O'Berry
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Ingrid Camila Possa-Paranhos
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Jared Butts
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Naraen Palanikumar
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Patrick Sweeney
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
34
|
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
|
35
|
Wei R, Li D, Jia S, Chen Y, Wang J. MC4R in Central and Peripheral Systems. Adv Biol (Weinh) 2023; 7:e2300035. [PMID: 37043700 DOI: 10.1002/adbi.202300035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/25/2023] [Indexed: 04/14/2023]
Abstract
Obesity has emerged as a critical and urgent health burden during the current global pandemic. Among multiple genetic causes, melanocortin receptor-4 (MC4R), involved in food intake and energy metabolism regulation through various signaling pathways, has been reported to be the lead genetic factor in severe and early onset obesity and hyperphagia disorders. Most previous studies have illustrated the roles of MC4R signaling in energy intake versus expenditure in the central system, while some evidence indicates that MC4R is also expressed in peripheral systems, such as the gut and endocrine organs. However, its physiopathological function remains poorly defined. This review aims to depict the central and peripheral roles of MC4R in energy metabolism and endocrine hormone homeostasis, the diversity of phenotypes, biased downstream signaling caused by distinct MC4R mutations, and current drug development targeting the receptor.
Collapse
Affiliation(s)
- Ran Wei
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Sheng Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| |
Collapse
|
36
|
Horwitz A, Birk R. Adipose Tissue Hyperplasia and Hypertrophy in Common and Syndromic Obesity-The Case of BBS Obesity. Nutrients 2023; 15:3445. [PMID: 37571382 PMCID: PMC10421039 DOI: 10.3390/nu15153445] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/16/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a metabolic state generated by the expansion of adipose tissue. Adipose tissue expansion depends on the interplay between hyperplasia and hypertrophy, and is mainly regulated by a complex interaction between genetics and excess energy intake. However, the genetic regulation of adipose tissue expansion is yet to be fully understood. Obesity can be divided into common multifactorial/polygenic obesity and monogenic obesity, non-syndromic and syndromic. Several genes related to obesity were found through studies of monogenic non-syndromic obesity models. However, syndromic obesity, characterized by additional features other than obesity, suggesting a more global role of the mutant genes related to the syndrome and, thus, an additional peripheral influence on the development of obesity, were hardly studied to date in this regard. This review summarizes present knowledge regarding the hyperplasia and hypertrophy of adipocytes in common obesity. Additionally, we highlight the scarce research on syndromic obesity as a model for studying adipocyte hyperplasia and hypertrophy, focusing on Bardet-Biedl syndrome (BBS). BBS obesity involves central and peripheral mechanisms, with molecular and mechanistic alternation in adipocyte hyperplasia and hypertrophy. Thus, we argue that using syndromic obesity models, such as BBS, can further advance our knowledge regarding peripheral adipocyte regulation in obesity.
Collapse
Affiliation(s)
| | - Ruth Birk
- Department of Nutrition, Faculty of Health Sciences, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
37
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Competition between stochastic neuropeptide signals calibrates the rate of satiation. RESEARCH SQUARE 2023:rs.3.rs-3185572. [PMID: 37546985 PMCID: PMC10402269 DOI: 10.21203/rs.3.rs-3185572/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
We investigated how transmission of hunger- and satiety-promoting neuropeptides, NPY and αMSH, is integrated at the level of intracellular signaling to control feeding. Receptors for these peptides use the second messenger cAMP. How cAMP integrates opposing peptide signals to regulate energy balance, and the in vivo spatiotemporal dynamics of endogenous peptidergic signaling, remain largely unknown. We show that AgRP axon stimulation in the paraventricular hypothalamus evokes probabilistic NPY release that triggers stochastic cAMP decrements in downstream MC4R-expressing neurons (PVHMC4R). Meanwhile, POMC axon stimulation triggers stochastic, αMSH-dependent cAMP increments. Release of either peptide impacts a ~100 μm diameter region, and when these peptide signals overlap, they compete to control cAMP. The competition is reflected by hunger-state-dependent differences in the amplitude and persistence of cAMP transients: hunger peptides are more efficacious in the fasted state, satiety peptides in the fed state. Feeding resolves the competition by simultaneously elevating αMSH release and suppressing NPY release, thereby sustaining elevated cAMP in PVHMC4R neurons. In turn, cAMP potentiates feeding-related excitatory inputs and promotes satiation across minutes. Our findings highlight how biochemical integration of opposing, quantal peptide signals during energy intake orchestrates a gradual transition between stable states of hunger and satiety.
Collapse
Affiliation(s)
- Stephen X Zhang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Co-corresponding authors
| | - Angela Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Paula K Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lauren F Christenson
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Present address: Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter N Kalugin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Akash Pal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
- Co-corresponding authors
| |
Collapse
|
38
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Competition between stochastic neuropeptide signals calibrates the rate of satiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548551. [PMID: 37503012 PMCID: PMC10369917 DOI: 10.1101/2023.07.11.548551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
We investigated how transmission of hunger- and satiety-promoting neuropeptides, NPY and αMSH, is integrated at the level of intracellular signaling to control feeding. Receptors for these peptides use the second messenger cAMP, but the messenger's spatiotemporal dynamics and role in energy balance are controversial. We show that AgRP axon stimulation in the paraventricular hypothalamus evokes probabilistic and spatially restricted NPY release that triggers stochastic cAMP decrements in downstream MC4R-expressing neurons (PVH MC4R ). Meanwhile, POMC axon stimulation triggers stochastic, αMSH-dependent cAMP increments. NPY and αMSH competitively control cAMP, as reflected by hunger-state-dependent differences in the amplitude and persistence of cAMP transients evoked by each peptide. During feeding bouts, elevated αMSH release and suppressed NPY release cooperatively sustain elevated cAMP in PVH MC4R neurons, thereby potentiating feeding-related excitatory inputs and promoting satiation across minutes. Our findings highlight how state-dependent integration of opposing, quantal peptidergic events by a common biochemical target calibrates energy intake.
Collapse
|
39
|
Popov MY, Lepik OV, Kozlovskii VL, Popov YV. Pharmacological strategies for appetite modulation in eating disorders: a narrative review. CONSORTIUM PSYCHIATRICUM 2023; 4:79-90. [PMID: 38250648 PMCID: PMC10795951 DOI: 10.17816/cp6150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/15/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND A substantial increase in the prevalence of eating disorders has been noticed over the past decades. Priority in the treatment of eating disorders is justifiably given to psychosocial interventions. However, it is also well known that centrally acting drugs can significantly affect appetite and food consumption. AIM To narratively review the available neurobiological data on the mechanisms of central regulation of eating behavior as a rationale to summarize pharmacological strategies for appetite modulation in eating disorders. METHODS The authors have carried out a narrative review of scientific papers published from January 2013 to March 2023 in the PubMed and Web of Science electronic databases. Studies were considered eligible if they included data on the neurobiological mechanisms of appetite regulation or the results of clinical trials of centrally acting drugs in eating disorders. Relevant studies were included regardless of their design. Descriptive analysis was used to summarize the obtained data. RESULTS The review included 51 studies. The available neurobiological and clinical data allowed us to identify the following pharmacological strategies for appetite modulation in eating disorders: serotonergic, catecholaminergic, amino acidergic and peptidergic. However, implementation of these data into clinical practice difficult due to an insufficient number of good-quality studies, which is particularly relevant for adolescents as there is a research gap in this population. CONCLUSION The progress in neurobiological understanding of the mechanisms of central regulation of appetite opens opportunities for new pharmacotherapeutic approaches aimed at changing the patterns of eating behavior. Obviously, treatment of eating disorders is a much broader problem and cannot be reduced to the correction of eating patterns. Nevertheless, at certain stages of treatment, drug-induced modulation of appetite can play an important role among multi-targeted biological and psychosocial interventions. Translation of neurobiological data into clinical practice requires a large number of clinical studies to confirm the long-term efficacy and safety of pharmacotherapeutic approaches and to develop personalized algorithms for the treatment of various forms of eating disorders in different age groups.
Collapse
Affiliation(s)
- Mikhail Y. Popov
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology
| | - Olga V. Lepik
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology
| | | | - Yuri V. Popov
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology
| |
Collapse
|
40
|
Becetti I, Bwenyi EL, de Araujo IE, Ard J, Cryan JF, Farooqi IS, Ferrario CR, Gluck ME, Holsen LM, Kenny PJ, Lawson EA, Lowell BB, Schur EA, Stanley TL, Tavakkoli A, Grinspoon SK, Singhal V. The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets: A Report from the 23rd Annual Harvard Nutrition Obesity Symposium. Am J Clin Nutr 2023; 118:314-328. [PMID: 37149092 PMCID: PMC10375463 DOI: 10.1016/j.ajcnut.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023] Open
Abstract
Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.
Collapse
Affiliation(s)
- Imen Becetti
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States.
| | - Esther L Bwenyi
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Jamy Ard
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Bariatric and Weight Management Center, Wake Forest Baptist Health, Winston-Salem, NC, United States; Center on Diabetes, Obesity, and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Hypertension and Vascular Research Center, Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Maya Angelou Center for Healthy Equity, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ismaa Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom; Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom; Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Carrie R Ferrario
- Department of Pharmacology, Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI, United States
| | - Marci E Gluck
- National Institutes of Health, Phoenix, AZ, United States; National Institute of Diabetes and Digestive and Kidney Disease, Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, Phoenix, AZ, United States
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, United States; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Elizabeth A Lawson
- Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Ellen A Schur
- Division of General Internal Medicine, University of Washington, Seattle, WA, United States; Univeristy of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States; Univeristy of Washington Nutrition and Obesity Research Center, University of Washington, Seattle, WA, United States; Clinical and Translational Research Services Core, University of Washington, Seattle, WA, United States
| | - Takara L Stanley
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ali Tavakkoli
- Division of General and Gastrointestinal (GI) Surgery, Center for Weight Management and Wellness, Advanced Minimally Invasive Fellowship, Harvard Medical School, Boston, MA, United States
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Pediatric Endocrinology and Obesity Medicine, Massachusetts General Hospital, Boston, MA, United States; Pediatric Program MGH Weight Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
41
|
Xu Y, Jiang Z, Li H, Cai J, Jiang Y, Otiz-Guzman J, Xu Y, Arenkiel BR, Tong Q. Lateral septum as a melanocortin downstream site in obesity development. Cell Rep 2023; 42:112502. [PMID: 37171957 DOI: 10.1016/j.celrep.2023.112502] [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: 11/08/2022] [Revised: 03/27/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
The melanocortin pathway is well established to be critical for body-weight regulation in both rodents and humans. Despite extensive studies focusing on this pathway, the downstream brain sites that mediate its action are not clear. Here, we found that, among the known paraventricular hypothalamic (PVH) neuron groups, those expressing melanocortin receptors 4 (PVHMc4R) preferably project to the ventral part of the lateral septum (LSv), a brain region known to be involved in emotional behaviors. Photostimulation of PVHMc4R neuron terminals in the LSv reduces feeding and causes aversion, whereas deletion of Mc4Rs or disruption of glutamate release from LSv-projecting PVH neurons causes obesity. In addition, disruption of AMPA receptor function in PVH-projected LSv neurons causes obesity. Importantly, chronic inhibition of PVH- or PVHMc4R-projected LSv neurons causes obesity associated with reduced energy expenditure. Thus, the LSv functions as an important node in mediating melanocortin action on body-weight regulation.
Collapse
Affiliation(s)
- Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hongli Li
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanyan Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joshua Otiz-Guzman
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Liu H, Li X, Li P, Hai R, Li J, Fan Q, Wang X, Chen Y, Cao X, Zhang X, Gao R, Wang K, Du C. Glutamatergic melanocortin-4 receptor neurons regulate body weight. FASEB J 2023; 37:e22920. [PMID: 37078546 DOI: 10.1096/fj.202201786r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/22/2023] [Accepted: 03/30/2023] [Indexed: 04/21/2023]
Abstract
The locus coeruleus (LC), enriched in vesicular glutamate transporter 2 (VGlut2) neurons, is a potential homeostasis-regulating hub. However, the identity of melanocortin-4 receptor (MC4R) neurons in the paraventricular nucleus (PVN) of the hypothalamus, PVNVGlut2::MC4R and LCVGlut2::MC4R regulation of body weight, and axonal projections of LCVGlut2 neurons remain unclear. Conditional knockout of MC4R in chimeric mice was used to confirm the effects of VGlut2. Interscapular brown adipose tissue was injected with pseudorabies virus to study the central nervous system projections. We mapped the LCVGlut2 circuitry. Based on the Cre-LoxP recombination system, specific knockdown of MC4R in VGlut2 neurons resulted in weight gain in chimeric mice. Adeno-associated virus-mediated knockdown of MC4R expression in the PVN and LC had potential superimposed effects on weight gain, demonstrating the importance of VGlut2 neurons. Unlike these wide-ranging efferent projections, the PVN, hypothalamic arcuate nucleus, supraoptic nucleus of the lateral olfactory tegmental nuclei, and nucleus tractus solitarius send excitatory projections to LCVGlut2 neurons. The PVN → LC glutamatergic MC4R long-term neural circuit positively affected weight management and could help treat obesity.
Collapse
Affiliation(s)
- Haodong Liu
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaojing Li
- College of Horticulture and Plant Protection, Inner Mongolia Agricultural University, Hohhot, China
| | - Penghui Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Rihan Hai
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Jiacheng Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Qi Fan
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xing Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Yujie Chen
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Xiaojuan Cao
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Xiaoyu Zhang
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Ruifeng Gao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Kun Wang
- Institute of Cereal and Oil Crops, Hebei Academy of Agricultural and Forestry Sciences, Shijiazhuang, China
- Key Laboratory of Crop Cultivation Physiology and Green Production in Hebei Province, Shijiazhuang, China
| | - Chenguang Du
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| |
Collapse
|
43
|
Semple EA, Harberson MT, Xu B, Rashleigh R, Cartwright TL, Braun JJ, Custer AC, Liu C, Hill JW. Melanocortin 4 receptor signaling in Sim1 neurons permits sexual receptivity in female mice. Front Endocrinol (Lausanne) 2023; 14:983670. [PMID: 37033219 PMCID: PMC10080118 DOI: 10.3389/fendo.2023.983670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Female sexual dysfunction affects approximately 40% of women in the United States, yet few therapeutic options exist for these patients. The melanocortin system is a new treatment target for hypoactive sexual desire disorder (HSDD), but the neuronal pathways involved are unclear. Methods In this study, the sexual behavior of female MC4R knockout mice lacking melanocortin 4 receptors (MC4Rs) was examined. The mice were then bred to express MC4Rs exclusively on Sim1 neurons (tbMC4RSim1 mice) or on oxytocin neurons (tbMC4ROxt mice) to examine the effect on sexual responsiveness. Results MC4R knockout mice were found to approach males less and have reduced receptivity to copulation, as indicated by a low lordosis quotient. These changes were independent of body weight. Lordosis behavior was normalized in tbMC4RSim1 mice and improved in tbMC4ROxt mice. In contrast, approach behavior was unchanged in tbMC4RSim1 mice but greatly increased in tbMC4ROxt animals. The changes were independent of melanocortin-driven metabolic effects. Discussion These results implicate MC4R signaling in Oxt neurons in appetitive behaviors and MC4R signaling in Sim1 neurons in female sexual receptivity, while suggesting melanocortin-driven sexual function does not rely on metabolic neural circuits.
Collapse
Affiliation(s)
- Erin A. Semple
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Mitchell T. Harberson
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Baijie Xu
- Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, United States
| | - Rebecca Rashleigh
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Tori L. Cartwright
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Jessica J. Braun
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Amy C. Custer
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Chen Liu
- Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, United States
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, United States
| |
Collapse
|
44
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
45
|
Wagner S, Brierley DI, Leeson-Payne A, Jiang W, Chianese R, Lam BYH, Dowsett GKC, Cristiano C, Lyons D, Reimann F, Gribble FM, Martinez de Morentin PB, Yeo GSH, Trapp S, Heisler LK. Obesity medication lorcaserin activates brainstem GLP-1 neurons to reduce food intake and augments GLP-1 receptor agonist induced appetite suppression. Mol Metab 2023; 68:101665. [PMID: 36592795 PMCID: PMC9841057 DOI: 10.1016/j.molmet.2022.101665] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Overweight and obesity are endemic in developed countries, with a substantial negative impact on human health. Medications developed to treat obesity include agonists for the G-protein coupled receptors glucagon-like peptide-1 (GLP-1R; e.g. liraglutide), serotonin 2C (5-HT2CR; e.g, lorcaserin), and melanocortin4 (MC4R) which reduce body weight primarily by suppressing food intake. However, the mechanisms underlying the therapeutic food intake suppressive effects are still being defined and were investigated here. METHODS We profiled PPG neurons in the nucleus of the solitary tract (PPGNTS) using single nucleus RNA sequencing (Nuc-Seq) and histochemistry. We next examined the requirement of PPGNTS neurons for obesity medication effects on food intake by virally ablating PPGNTS neurons. Finally, we assessed the effects on food intake of the combination of liraglutide and lorcaserin. RESULTS We found that 5-HT2CRs, but not GLP-1Rs or MC4Rs, were widespread in PPGNTS clusters and that lorcaserin significantly activated PPGNTS neurons. Accordingly, ablation of PPGNTS neurons prevented the reduction of food intake by lorcaserin but not MC4R agonist melanotan-II, demonstrating the functional significance of PPGNTS 5-HT2CR expression. Finally, the combination of lorcaserin with GLP-1R agonists liraglutide or exendin-4 produced greater food intake reduction as compared to either monotherapy. CONCLUSIONS These findings identify a necessary mechanism through which obesity medication lorcaserin produces its therapeutic benefit, namely brainstem PPGNTS neurons. Moreover, these data reveal a strategy to augment the therapeutic profile of the current frontline treatment for obesity, GLP-1R agonists, via coadministration with 5-HT2CR agonists.
Collapse
Affiliation(s)
- Stefan Wagner
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Daniel I Brierley
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | | | - Wanqing Jiang
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, 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
| | - Georgina K C Dowsett
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | | | - David Lyons
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Frank Reimann
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Fiona M Gribble
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, 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
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK.
| | - Lora K Heisler
- The Rowett Institute, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
46
|
Bernard A, Ojeda Naharros I, Yue X, Mifsud F, Blake A, Bourgain-Guglielmetti F, Ciprin J, Zhang S, McDaid E, Kim K, Nachury MV, Reiter JF, Vaisse C. MRAP2 regulates energy homeostasis by promoting primary cilia localization of MC4R. JCI Insight 2023; 8:e155900. [PMID: 36692018 PMCID: PMC9977312 DOI: 10.1172/jci.insight.155900] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 11/30/2022] [Indexed: 01/24/2023] Open
Abstract
The G protein-coupled receptor melanocortin-4 receptor (MC4R) and its associated protein melanocortin receptor-associated protein 2 (MRAP2) are essential for the regulation of food intake and body weight in humans. MC4R localizes and functions at the neuronal primary cilium, a microtubule-based organelle that senses and relays extracellular signals. Here, we demonstrate that MRAP2 is critical for the weight-regulating function of MC4R neurons and the ciliary localization of MC4R. More generally, our study also reveals that GPCR localization to primary cilia can require specific accessory proteins that may not be present in heterologous cell culture systems. Our findings further demonstrate that targeting of MC4R to neuronal primary cilia is essential for the control of long-term energy homeostasis and suggest that genetic disruption of MC4R ciliary localization may frequently underlie inherited forms of obesity.
Collapse
Affiliation(s)
| | | | - Xinyu Yue
- Department of Medicine and The Diabetes Center
| | | | - Abbey Blake
- Department of Medicine and The Diabetes Center
| | | | | | - Sumei Zhang
- Department of Medicine and The Diabetes Center
| | - Erin McDaid
- Department of Medicine and The Diabetes Center
| | - Kellan Kim
- Department of Medicine and The Diabetes Center
| | | | - Jeremy F. Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | | |
Collapse
|
47
|
Murphy S, Collis Glynn M, Dixon TN, Grill HJ, McNally GP, Ong ZY. Nucleus of the solitary tract A2 neurons control feeding behaviors via projections to the paraventricular hypothalamus. Neuropsychopharmacology 2023; 48:351-361. [PMID: 36114285 PMCID: PMC9751294 DOI: 10.1038/s41386-022-01448-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 12/26/2022]
Abstract
Hindbrain NTS neurons are highly attuned to internal physiological and external environmental factors that contribute to the control of food intake but the relevant neural phenotypes and pathways remain elusive. Here, we investigated the role of NTS A2 neurons and their projections in the control of feeding behaviors. In male TH Cre rats, we first confirmed selective targeting of NTS A2 neurons and showed that chemogenetic stimulation of these neurons significantly suppressed dark cycle food intake, deprivation re-feed and high fat diet intake. Despite reducing intake, activation of NTS A2 neurons had no effect on food approach, anxiety-like behaviors, locomotor activity, blood glucose levels nor did it induce nausea/malaise, thus revealing a selective role for these neurons in the consummatory aspect of food intake control. Pathway-specific mapping and manipulation of NTS A2 neurons showed that these effects were mediated by NTS A2 neurons projecting to the paraventricular nucleus of the hypothalamus (PVH) because chemogenetic activation of these projections, but not projections to bed nucleus of the stria terminalis (BNST), reduced food intake. Cell-type specific analyses demonstrated that activation of NTS A2 neurons recruited both PVH oxytocin (OT)- and corticotropin-releasing factor (CRF)-expressing neurons, and plasma analyses showed increased plasma corticosterone following NTS A2 stimulation. While we also showed that chemogenetic inhibition of NTS A2 neurons attenuated the intake inhibitory effects of CCK, the specificity of transgene expression was low. Together, these findings showed that NTS A2 neurons are sufficient to control the consummatory aspects of feeding, regardless of energy status or food palatability and identified their projections to PVH, but not BNST, in food intake control.
Collapse
Affiliation(s)
- Stephanie Murphy
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Metika Collis Glynn
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Tiarani N Dixon
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gavan P McNally
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Zhi Yi Ong
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia.
| |
Collapse
|
48
|
Hajdarovic KH, Yu D, Webb AE. Understanding the aging hypothalamus, one cell at a time. Trends Neurosci 2022; 45:942-954. [PMID: 36272823 PMCID: PMC9671837 DOI: 10.1016/j.tins.2022.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022]
Abstract
The hypothalamus is a brain region that integrates signals from the periphery and the environment to maintain organismal homeostasis. To do so, specialized hypothalamic neuropeptidergic neurons control a range of processes, such as sleep, feeding, the stress response, and hormone release. These processes are altered with age, which can affect longevity and contribute to disease status. Technological advances, such as single-cell RNA sequencing, are upending assumptions about the transcriptional identity of cell types in the hypothalamus and revealing how distinct cell types change with age. In this review, we summarize current knowledge about the contribution of hypothalamic functions to aging. We highlight recent single-cell studies interrogating distinct cell types of the mouse hypothalamus and suggest ways in which single-cell 'omics technologies can be used to further understand the aging hypothalamus and its role in longevity.
Collapse
Affiliation(s)
| | - Doudou Yu
- Graduate program in Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
49
|
Chang HC, Lee YJ, Javed S, Haque M, Chang YT, Lin YC, Oram C, Huang WH. rAAV-CRISPRa therapy corrects Rai1 haploinsufficiency and rescues selective disease features in Smith-Magenis syndrome mice. J Biol Chem 2022; 299:102728. [PMID: 36410433 PMCID: PMC9762195 DOI: 10.1016/j.jbc.2022.102728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/20/2022] Open
Abstract
Haploinsufficiency in retinoic acid induced 1 (RAI1) causes Smith-Magenis syndrome (SMS), a severe neurodevelopmental disorder characterized by neurocognitive deficits and obesity. Currently, curative treatments for SMS do not exist. Here, we take a recombinant adeno-associated virus (rAAV)-clustered regularly interspaced short palindromic repeats activation (CRISPRa) approach to increase expression of the remaining intact Rai1 allele. Building upon our previous work that found the paraventricular nucleus of hypothalamus plays a central role in SMS pathogenesis, we performed paraventricular nucleus of hypothalamus-specific rAAV-CRISPRa therapy by increasing endogenous Rai1 expression in SMS (Rai1±) mice. We found that rAAV-CRISPRa therapy rescues excessive repetitive behavior, delays the onset of obesity, and partially reduces hyperphagia in SMS mice. Our work provides evidence that rAAV-CRISPRa therapy during early adolescence can boost the expression of healthy Rai1 allele and modify disease progression in a mouse model of Smith-Magenis syndrome.
Collapse
Affiliation(s)
- Hao-Cheng Chang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu-Ju Lee
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Sehrish Javed
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Minza Haque
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Ya-Ting Chang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu Cheng Lin
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Cameron Oram
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Wei-Hsiang Huang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Québec, Canada,Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada,For correspondence: Wei-Hsiang Huang
| |
Collapse
|
50
|
Autry AE. Function of brain-derived neurotrophic factor in the hypothalamus: Implications for depression pathology. Front Mol Neurosci 2022; 15:1028223. [PMID: 36466807 PMCID: PMC9708894 DOI: 10.3389/fnmol.2022.1028223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Depression is a prevalent mental health disorder and is the number one cause of disability worldwide. Risk factors for depression include genetic predisposition and stressful life events, and depression is twice as prevalent in women compared to men. Both clinical and preclinical research have implicated a critical role for brain-derived neurotrophic factor (BDNF) signaling in depression pathology as well as therapeutics. A preponderance of this research has focused on the role of BDNF and its primary receptor tropomyosin-related kinase B (TrkB) in the cortex and hippocampus. However, much of the symptomatology for depression is consistent with disruptions in functions of the hypothalamus including changes in weight, activity levels, responses to stress, and sociability. Here, we review evidence for the role of BDNF and TrkB signaling in the regions of the hypothalamus and their role in these autonomic and behavioral functions associated with depression. In addition, we identify areas for further research. Understanding the role of BDNF signaling in the hypothalamus will lead to valuable insights for sex- and stress-dependent neurobiological underpinnings of depression pathology.
Collapse
Affiliation(s)
- Anita E. Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
- *Correspondence: Anita E. Autry,
| |
Collapse
|