1
|
Sabeti Akbar-Abad M, Majidpour M, Keykha F, Maleki M, Piroozan Y, Saravani R, Zandhaghighi M, Shahriari H, Sargazi S. Preliminary insight into the potential role of Leptin Receptor Polymorphisms in Type 2 Diabetes Risk: case-control study and bioinformatics analysis. J Diabetes Metab Disord 2025; 24:113. [PMID: 40331155 PMCID: PMC12049348 DOI: 10.1007/s40200-025-01617-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025]
Abstract
Background Type 2 diabetes mellitus (T2DM) develops primarily from obesity as leptin (LEP) functions as an essential adipokine that controls metabolic regulation, energy balance activities, and glucose maintenance. The T2DM and obesity susceptibility traits are believed to be affected by genetic variations in the leptin receptor gene (LEPR), disrupting LEP signaling mechanisms. This case-control study investigates the association of these variants with T2DM risk in a Southeastern Iranian population. Methods A case-control study was conducted involving 450 T2DM patients and 450 matched healthy controls from Zahedan. Genomic DNA for this study was isolated from peripheral blood samples, and genotyping for the specified LEPR rs1137100, rs1137101, and rs1805094 polymorphisms was conducted using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Computational analysis created a gene-gene interaction network, highlighting LEPR as a central hub gene and detailing its interactions with related genes. Results Genetic models, such as codominant heterozygous (p-value = 0.009), dominant (p-value = 0.006), recessive (p-value = 0.008), and allelic (p-value = 0.011), all showed that the rs1137100 (A/G) polymorphism lowered the risk of T2DM. Several genetic models linked polymorphisms at the rs1137101 (G/A) and rs1805094 (G/C) loci to a higher risk of T2DM: The genetic models that were looked at were polymorphism rs1137101 (G/A) in codominant Homozygous (p-value = 0.031) and recessive (p-value = 0.028), as well as polymorphism rs1805094 (G/C) in codominant heterozygous (p-value = 0.009), dominant (p-value = 0.001), excess (p-value = 0.008), and allelic (p-value = 0.001). The research demonstrated a profound linkage disequilibrium (LD) among studied variants, especially in the LEPR haplotypes and across various blocks, with differing levels of association strength. The gene-gene interaction network for the LEPR gene highlights its strong associations with several key regulatory genes: LEP, PTPN11, STAT3, POMC, JAK2, IL6, and SOCS3. Conclusion We found a significant correlation between LEPR gene polymorphisms and the risk of T2DM, highlighting the prominent role of genetic factors in developing such a metabolic disorder. By elucidating the association between LEPR variations and susceptibility to T2DM, our findings enhance the understanding of molecular mechanisms involved in endocrine dysregulation and highlight the importance of including genetic profiling in clinical practice.
Collapse
Affiliation(s)
- Mahboobeh Sabeti Akbar-Abad
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fatemeh Keykha
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Mohsen Maleki
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Yegane Piroozan
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mehdi Zandhaghighi
- Department of Medical Microbiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Infectious Diseases and Tropical Medicine Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saman Sargazi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
2
|
Yoon JS, Gamu D, Gibson WT, Lynn FC. NPAS4 Depletion in POMC Neurons Protects From Obesity and Alters the Feeding-regulated Transcriptome in Male Mice. Endocrinology 2025; 166:bqaf083. [PMID: 40296822 PMCID: PMC12123070 DOI: 10.1210/endocr/bqaf083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/11/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Immediate early genes (IEGs), such as neuronal PAS domain protein 4 (Npas4), are induced as part of the response to environmental stimuli. In the arcuate nucleus (ARC), proopiomelanocortin (POMC) neurons are critical in detecting peripheral signals to regulate food intake. To date, Npas4 has not been studied in the context of regulating food intake, and its sites of action in the ARC are unknown. We found that Npas4 was induced in POMC neurons by refeeding, oral glucose, and a high-fat diet (HFD). In order to explore the role of NPAS4 in POMC neurons, a conditional knockout approach was used. Male mice with Npas4 knockout in POMC neurons showed significantly reduced body weight starting at 10 weeks of HFD, which was due to decreased food intake. Single-cell RNA sequencing on ARC cells demonstrated that POMC neurons of knockout mice showed an enhanced refeeding-induced transcriptional response, dysregulated IEG expression in response to refeeding, and reduced expression of genes encoding gamma-aminobutyric acid (GABA)-A receptor subunits. Cell-to-cell communication analysis revealed that POMC neurons of knockout mice lost inhibitory GABAergic signaling inputs and gained excitatory glutamatergic signaling inputs. Taken together, these results suggest that Npas4 tempers the activity of POMC neurons and loss of Npas4 causes impairments in nutrient intake sensing. Mechanistically, this results from reduced expression of inhibitory GABA-A receptors and an overall increase in the feeding-induced POMC neuron transcriptional response. In conclusion, we report a role for the transcription factor Npas4 in POMC neurons of the ARC and demonstrate its importance in controlling feeding behavior in states of overnutrition.
Collapse
Affiliation(s)
- Ji Soo Yoon
- BC Children's Hospital Research Institute, Vancouver, BC, Canada V5Z 4H4
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| | - Daniel Gamu
- BC Children's Hospital Research Institute, Vancouver, BC, Canada V5Z 4H4
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| | - William T Gibson
- BC Children's Hospital Research Institute, Vancouver, BC, Canada V5Z 4H4
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Vancouver, BC, Canada V5Z 4H4
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| |
Collapse
|
3
|
Chen D, Yang YY, Yang Y. Astrocyte Loss Augments Body Weight Through Reduction in Adipose Sympathetic Outflows. Glia 2025; 73:1068-1076. [PMID: 39780483 PMCID: PMC11922664 DOI: 10.1002/glia.24673] [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: 08/14/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Emerging evidence indicates that astrocytes modulate energy metabolism and homeostasis. However, one important but poorly understood element is the necessity of astrocytes in the control of body weight. Here, we apply viral vector-assisted brain-region selective loss of astrocytes to define physiological roles played by astrocytes in the arcuate nucleus of the hypothalamus (ARH) and to elucidate the involved mechanism. We find that astrocyte loss potently augments body weight in adult mice fed chow or high-fat diet. Mechanistically, we find that the loss of astrocytes reduces adipose tissue norepinephrine (NE) contents and chemogenetic stimulation of adipose tissue sympathetic inputs reverses the astrocyte loss-induced increase in body weight. Collectively, our findings in this study suggest a crucial physiological role of astrocytes in preventing diet-induced energy surfeit and obesity by modulating adipose tissue lipid metabolism through central sympathetic outflows to adipose tissues.
Collapse
Affiliation(s)
- Dan Chen
- Department of Medicine Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | - Yunlei Yang
- Department of Medicine Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
4
|
Kaneko K, Lu W, Xu Y, Morozov A, Fukuda M. The small GTPase Rap1 in POMC neurons regulates leptin actions and glucose metabolism. Mol Metab 2025; 95:102117. [PMID: 40024570 PMCID: PMC11938153 DOI: 10.1016/j.molmet.2025.102117] [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: 12/06/2024] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
The hypothalamic leptin-proopiomelanocortin (POMC) pathway is critical for regulating metabolism. POMC neurons in the arcuate nucleus respond to leptin and play a pivotal role in mediating energy and glucose balance. However, during diet-induced obesity (DIO), these neurons often develop resistance to exogenous leptin. Recently, the small GTPase Rap1 has been implicated as an inhibitor of neuronal leptin signaling; however, its specific role within POMC neurons remains unexplored. We generated tamoxifen-inducible, POMC neuron-specific Rap1 knockout mice to selectively delete both Rap1a and Rap1b isoforms in POMC neurons. By analyzing these mice through metabolic phenotyping, immunohistochemistry, and biochemical assays, we show that deleting Rap1a and Rap1b in POMC neurons prior to exposing the mice to a high-fat diet significantly prevented weight gain compared to control mice. Furthermore, while DIO mice with intact Rap1 failed to respond to exogenous leptin, genetically removing the Rap1 genes from DIO mice enhanced the ability of exogenous leptin to induce anorectic effects. Remarkably, acute deletion of Rap1 in POMC neurons of already obese mice improved hyperglycemia within one week, with minimal effect on body weight. This glycemic improvement was accompanied by improved glucose tolerance, enhanced insulin sensitivity, and improved cellular insulin signaling. Collectively, these findings suggest that loss of Rap1 in POMC neurons enhances leptin sensitivity, acutely improves glucose balance, and may offer a potential strategy to lower hyperglycemia in dietary obesity.
Collapse
Affiliation(s)
- Kentaro Kaneko
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kanagawa, Japan
| | - Weisheng Lu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Alexei Morozov
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, 24016, USA
| | - Makoto Fukuda
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Jiao Z, Gao T, Wang X, Wang A, Ma Y, Feng L, Gao L, Gou L, Zhang W, Biglari N, Boxer EE, Steuernagel L, Ding X, Yu Z, Li M, Gao M, Hao M, Zhou H, Cao X, Li S, Jiang T, Qi J, Jia X, Feng Z, Ren B, Chen Y, Shi X, Wang D, Wang X, Han L, Liang Y, Qian L, Jin C, Huang J, Deng W, Wang C, Li E, Hu Y, Tao Z, Li H, Yu X, Xu M, Chang HC, Zhang Y, Xu H, Yan J, Li A, Luo Q, Stoop R, Sternson SM, Brüning JC, Anderson DJ, Poo MM, Sun Y, Xu S, Gong H, Sun YG, Xu X. Projectome-based characterization of hypothalamic peptidergic neurons in male mice. Nat Neurosci 2025; 28:1073-1088. [PMID: 40140607 DOI: 10.1038/s41593-025-01919-0] [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: 04/27/2023] [Accepted: 02/07/2025] [Indexed: 03/28/2025]
Abstract
The hypothalamus coordinately regulates physiological homeostasis and innate behaviors, yet the detailed arrangement of hypothalamic axons remains unclear. Here we mapped the whole-brain projections of over 7,000 hypothalamic neurons expressing distinct neuropeptides in male mice, identifying 2 main classes and 31 types using single-neuron projectome analysis. These classes/types exhibited regionally biased soma distribution and specific neuropeptide enrichment. Notably, many projectome types extended long-range axon collaterals to distinct brain regions, allowing single axons to co-regulate multiple targets. We uncovered topographic organization of certain peptidergic axons at specific targets, along with diverse single-neuron projectome patterns in Orexin, Agrp and Pomc populations. Furthermore, hypothalamic peptidergic neurons showed correlated innervation of subdomains in the periaqueductal gray and organized into modular subnetworks within the hypothalamus, providing a structural basis for coordinated outputs. This dataset highlights the complexity of hypothalamic axonal projections and lays a foundation for future investigation of the circuit mechanisms underlying hypothalamic functions.
Collapse
Affiliation(s)
- Zhuolei Jiao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Taosha Gao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaofei Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ao Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yawen Ma
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Li Feng
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Le Gao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lingfeng Gou
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wen Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nasim Biglari
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Policlinic for Endocrinology, Diabetology and Preventive Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Emma E Boxer
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Howard Hughes Medical Institute; Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, USA
| | - Lukas Steuernagel
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Policlinic for Endocrinology, Diabetology and Preventive Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Xiaojing Ding
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zixian Yu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mingjuan Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengtong Gao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Mingkun Hao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Hua Zhou
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xuanzi Cao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Shuaishuai Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Jiamei Qi
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Xueyan Jia
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Zhao Feng
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Biyu Ren
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yu Chen
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxue Shi
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Dan Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xinran Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Luyao Han
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yikai Liang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Liuqin Qian
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chenxi Jin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiawen Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wei Deng
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Congcong Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - E Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yue Hu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zi Tao
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Humingzhu Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiang Yu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences and Peking University McGovern Institute, Peking University, Beijing, China
| | - Min Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Hung-Chun Chang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yifeng Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Huatai Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jun Yan
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Anan Li
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Qingming Luo
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Ron Stoop
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital Center (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Scott M Sternson
- Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA, USA
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Policlinic for Endocrinology, Diabetology and Preventive Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - David J Anderson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Howard Hughes Medical Institute; Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, USA
| | - Mu-Ming Poo
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Shengjing Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Hui Gong
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China.
| | - Yan-Gang Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Xiaohong Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Slominski RM, Raman C, Jetten AM, Slominski AT. Neuro-immuno-endocrinology of the skin: how environment regulates body homeostasis. Nat Rev Endocrinol 2025:10.1038/s41574-025-01107-x. [PMID: 40263492 DOI: 10.1038/s41574-025-01107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/24/2025]
Abstract
The skin, including the hypodermis, is the largest organ of the body. The epidermis, the uppermost layer, is in direct contact with the environment and is exposed to environmental stressors, including solar radiation and biological, chemical and physical factors. These environmental factors trigger local responses within the skin that modulate homeostasis on both the cutaneous and systemic levels. Using mediators in common with brain pathways, immune and neuroendocrine systems within the skin regulate these responses to activate various signal transduction pathways and influence the systemic endocrine and immune systems in a context-dependent manner. This skin neuro-immuno-endocrine system is compartmentalized through the formation of epidermal, dermal, hypodermal and adnexal regulatory units. These units can act separately or in concert to preserve skin integrity, allow for adaptation to a changing environment and prevent the development of pathological processes. Through activation of peripheral nerve endings, the release of neurotransmitters, hormones, neuropeptides, and cytokines and/or chemokines into the circulation, or by priming circulating and resident immune cells, this system affects central coordinating centres and global homeostasis, thus adjusting the body's homeostasis and allostasis to optimally respond to the changing environment.
Collapse
Affiliation(s)
- Radomir M Slominski
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chander Raman
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL, USA.
- Veteran Administration Medical Center, Birmingham, AL, USA.
| |
Collapse
|
7
|
Llontop N, Mancilla C, Ojeda-Provoste P, Torres AK, Godoy A, Tapia-Rojas C, Kerr B. The methyl-CpG-binding protein 2 (Mecp2) regulates the hypothalamic mitochondrial function and white adipose tissue lipid metabolism. Life Sci 2025; 366-367:123478. [PMID: 39983816 DOI: 10.1016/j.lfs.2025.123478] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
OBJECTIVE The neuroepigenetic factor Mecp2 regulates gene expression and is thought to play a crucial role in energy homeostasis. Body weight is regulated at the hypothalamic level, where mitochondrial energy metabolism is necessary for its proper functioning, allowing the hypothalamus to respond to peripheral signals to maintain energy balance and modulate energy expenditure through the sympathetic nervous system. Since the mechanism by which genetic and environmental factors contribute to regulating energy balance is unclear, this study aims to understand the contribution of gene-environment interaction to maintaining energy balance and how its disruption alters hypothalamic cellular energy production, impacting the control of systemic metabolism. METHODS We used a mouse model of epigenetic disruption (Mecp2-null) to evaluate the impact of Mecp2 deletion on systemic and hypothalamic metabolism using physiological and cellular approaches. RESULTS Our study shows that the previously reported body weight gain in mice lacking the expression of Mecp2 is preceded by a hypothalamic mitochondrial dysfunction that disrupts hypothalamic function, leading to a dysfunctional communication between the hypothalamus and adipose tissue, thus impairing lipid metabolism. Our study has revealed three crucial aspects of the contribution of this critical epigenetic factor pivotal for a proper gene-environment interaction: i) Mecp2 drives a molecular mechanism to maintain cellular energy homeostasis, which is necessary for the proper functioning of the hypothalamus. ii) Mecp2 is necessary to maintain lipid metabolism in adipose tissue. iii) Mecp2 is a molecular bridge linking hypothalamic cellular energy metabolism and adipose tissue lipid metabolism. CONCLUSIONS Our results show that Mecp2 regulates the hypothalamic mitochondrial function and white adipose tissue lipid metabolism and probably alters the communication between these two tissues, which is critical for corporal energy homeostasis maintenance.
Collapse
Affiliation(s)
- Nuria Llontop
- Laboratory of Neuroendocrinology and Metabolism, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510157, Chile; Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510157, Chile
| | | | | | - Angie K Torres
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510157, Chile; Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, 8580702 Santiago, Chile
| | - Alejandro Godoy
- Laboratory of Endocrinology and Tumor Metabolism, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510157, Chile; Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510157, Chile; Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, 8580702 Santiago, Chile.
| | - Bredford Kerr
- Laboratory of Neuroendocrinology and Metabolism, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510157, Chile.
| |
Collapse
|
8
|
Stuber GD, Schwitzgebel VM, Lüscher C. The neurobiology of overeating. Neuron 2025:S0896-6273(25)00182-5. [PMID: 40185087 DOI: 10.1016/j.neuron.2025.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Food intake serves to maintain energy homeostasis; however, overeating can result in obesity, which is associated with serious health complications. In this review, we explore the intricate relationship between overeating, obesity, and the underlying neurobiological mechanisms. We review the homeostatic and hedonic feeding systems, highlighting the role of the hypothalamus and reward systems in controlling food intake and energy balance. Dysregulation in both these systems leads to overeating, as seen in genetic syndromes and environmental models affecting appetite regulation when consuming highly palatable food. The concept of "food addiction" is examined, drawing parallels to drug addiction. We discuss the cellular substrate for addiction-related behavior and current pharmacological obesity treatments-in particular, GLP-1 receptor agonists-showcasing synaptic plasticity in the context of overeating and palatable food exposure. A comprehensive model integrating insights from addiction research is proposed to guide effective interventions for maladaptive feeding behaviors. Ultimately, unraveling the neurobiological basis of overeating holds promise for addressing the pressing public health issue of obesity.
Collapse
Affiliation(s)
- Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Valerie M Schwitzgebel
- Pediatric Endocrinology and Diabetes Unit, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, 1211 Geneva, Switzerland; Institute of Genetics and Genomics (iGE3) in Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Christian Lüscher
- Institute of Genetics and Genomics (iGE3) in Geneva, University of Geneva, 1211 Geneva, Switzerland; Department of Basic Neurosciences, Medical Faculty, University of Geneva, 1211 Geneva, Switzerland; Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, 1211 Geneva, Switzerland; Synapsy Center for Mental Health Research, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
9
|
Zhuang S, Liu S, Li R, Duan H. Electroacupuncture alleviates insulin resistance and impacts the hypothalamic IRS-1/PI3K/AKT pathway and miRNA-29a-3p in a rat model of type 2 diabetes. Acupunct Med 2025; 43:104-113. [PMID: 40116412 DOI: 10.1177/09645284251327205] [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: 03/23/2025]
Abstract
OBJECTIVE This study aimed to explore the effects of electroacupuncture (EA) in a rat model of type 2 diabetes mellitus (T2DM) by examining the hypothalamic miRNA-29a-3p, insulin receptor substrate (IRS)-1 / phosphatidylinositol 3-kinase (PI3K) / protein kinase B (AKT) / signal transducer and activator of transcription (STAT)3 pathway, as well as metabolic molecules including proopiomelanocortin (POMC), neuropeptide Y (NPY) and agouti-related protein (AgRP), with the goal of revealing the molecular mechanisms by which EA mitigates T2DM. METHODS T2DM was induced by high-fat diet exposure followed by streptozotocin (STZ) injection, and LY294002 (a PI3K inhibitor) was administered to evaluate whether the effects of EA were mediated through the hypothalamic IRS-1/PI3K/AKT pathway. T2DM was induced in 36 rats that were assigned to model, EA and EA + LY294002 groups (n = 12 each). An additional 12 rats formed a healthy control group. Food intake, body weight and serum levels of fasting blood glucose (FBG), insulin, total cholesterol (TC), triglyceride (TG) and low-density lipoprotein (LDL) were measured. Hypothalamic micro (mi)RNA-29a-3p expression was detected by quantitative polymerase chain reaction (qPCR), while insulin receptor signaling pathway intermediates and metabolic molecules were analyzed using Western blotting, qPCR and immunohistochemistry. RESULTS After the EA intervention, T2DM rats showed a significant decrease in both food intake and body weight. In addition, there were reductions in serum concentrations of FBG, insulin, TC, TG and LDL. The homeostatic model assessment of insulin resistance (HOMA-IR) index also significantly declined. EA significantly decreased hypothalamic miRNA-29a-3p expression and enhanced IRS-1/PI3K/AKT/STAT3 pathway activity, while regulating POMC, NPY and AgRP expression. These effects of EA were at least partially reversed by LY294002 administration, which supports our hypothesis that EA mitigates T2DM via the hypothalamic IRS-1/PI3K/AKT pathway. CONCLUSION EA may improve hypothalamic IR by activating the IRS-1/PI3K/AKT/STAT3 pathway and regulating POMC, NPY and AgRP, thereby reducing food intake, correcting metabolic imbalance and mitigating T2DM. The effect of EA on the IRS-1/PI3K/AKT pathway may involve miRNA-29a-3p downregulation.
Collapse
Affiliation(s)
- Shuting Zhuang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Shaoyang Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Haoru Duan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Chen J, Cai M, Zhan C. Neuronal Regulation of Feeding and Energy Metabolism: A Focus on the Hypothalamus and Brainstem. Neurosci Bull 2025; 41:665-675. [PMID: 39704987 PMCID: PMC11978587 DOI: 10.1007/s12264-024-01335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/19/2024] [Indexed: 12/21/2024] Open
Abstract
In the face of constantly changing environments, the central nervous system (CNS) rapidly and accurately calculates the body's needs, regulates feeding behavior, and maintains energy homeostasis. The arcuate nucleus of the hypothalamus (ARC) plays a key role in this process, serving as a critical brain region for detecting nutrition-related hormones and regulating appetite and energy homeostasis. Agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons in the ARC are core elements that interact with other brain regions through a complex appetite-regulating network to comprehensively control energy homeostasis. In this review, we explore the discovery and research progress of AgRP neurons in regulating feeding and energy metabolism. In addition, recent advances in terms of feeding behavior and energy homeostasis, along with the redundant neural mechanisms involved in energy metabolism, are discussed. Finally, the challenges and opportunities in the field of neural regulation of feeding and energy metabolism are briefly discussed.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Meiting Cai
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Cheng Zhan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
11
|
Silva PIC, Pantoja LVPS, Conceição BC, Barbosa MEO, Soares LFR, Prediger RD, Fontes-Júnior EA, Freitas JJS, Maia CSF. Unraveling the Neuropharmacological Properties of Lippia alba: A Scientometric Approach. Pharmaceuticals (Basel) 2025; 18:420. [PMID: 40143196 PMCID: PMC11945933 DOI: 10.3390/ph18030420] [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: 12/27/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Lippia alba (Verbenaceae) is popularly known as lemon balm or false melissa and is one of the most widely used plants in traditional medicine in the Amazon region. In this study, we conducted a comprehensive bibliometric analysis, with conventional metrics associated with a critical review based on the neuropharmacological activities, to identify potential medical applications and also gaps in knowledge that require further investigation. Fifty-two articles were included according to the eligibility criteria. In the country analysis, Brazil emerged as the main contributor to research with the highest number of publications and citations. Notably, nine of the ten main research institutions are Brazilian, with the Universidade Federal de Santa Maria standing out with 761 citations. The keywords "anesthesia", "Lippia alba", and "essential oil" were the most frequent, highlighting their importance in this field. Essential oils are the most common type of extraction, which linalool, citral, geraniol, carvone, and limonene were the main constituents identified. According to the type of study, preclinical studies presented the highest frequency, primarily through fish experimental models. The main neuropharmacological activities identified were sedative-anesthetic, anxiolytic, anticonvulsant, and analgesic, with mechanisms of action via the GABAergic pathway. This bibliometric review provided new evidence reinforcing the potential of L. alba as a promising alternative for the treatment of neuropsychiatric disorders. It also highlighted existing knowledge gaps, mainly related to the comparison of the actions of the different chemotypes of the species and the investigation of the mechanisms underlying their neuropharmacological properties. Additionally, there is a lack of knowledge in other emerging areas related to the central nervous system, such as mood and cognitive disorders.
Collapse
Affiliation(s)
- Pedro I. C. Silva
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (P.I.C.S.); (L.V.P.S.P.); (B.C.C.)
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (M.E.O.B.); (L.F.R.S.); (E.A.F.-J.)
- Centro de Estudos Pré-Clínicos da Amazônia, Universidade do Estado do Pará, Belém 66087-662, PA, Brazil
| | - Lucas V. P. S. Pantoja
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (P.I.C.S.); (L.V.P.S.P.); (B.C.C.)
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Belém 66075-900, PA, Brazil
| | - Brenda C. Conceição
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (P.I.C.S.); (L.V.P.S.P.); (B.C.C.)
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Belém 66075-900, PA, Brazil
| | - Marta E. O. Barbosa
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (M.E.O.B.); (L.F.R.S.); (E.A.F.-J.)
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Belém 66075-900, PA, Brazil
| | - Luiza F. R. Soares
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (M.E.O.B.); (L.F.R.S.); (E.A.F.-J.)
| | - Rui Daniel Prediger
- Laboratório Experimental de Doenças Neurodegenerativas, Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis 88049-900, SC, Brazil;
| | - Enéas A. Fontes-Júnior
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (M.E.O.B.); (L.F.R.S.); (E.A.F.-J.)
| | - Jofre J. S. Freitas
- Centro de Estudos Pré-Clínicos da Amazônia, Universidade do Estado do Pará, Belém 66087-662, PA, Brazil
| | - Cristiane S. F. Maia
- Laboratório de Farmacologia da Inflamação e do Comportamento, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66075-900, PA, Brazil; (M.E.O.B.); (L.F.R.S.); (E.A.F.-J.)
| |
Collapse
|
12
|
Yan L, Zhang X, Jin L, Li Y, Chen Y, Zhang J, Sun Z, Qi J, Qu C, Dong G, Zhang Y, Jiang Q, Liu A, Li J. The ARC CRABP1 neurons play a crucial role in the regulation of energy homeostasis. Nat Commun 2025; 16:2319. [PMID: 40057489 PMCID: PMC11890859 DOI: 10.1038/s41467-025-57411-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 02/19/2025] [Indexed: 05/13/2025] Open
Abstract
Recent single-cell RNA sequencing study suggested that CRABP1 expressing neurons in the arcuate nucleus (ARCCRABP1 neurons) were a distinct group of neurons. However, the physiological role of ARCCRABP1 neurons remains unexplored. Here, we demonstrated that ARCCRABP1 neurons played a crucial role in regulation of energy homeostasis in male mice. Ablation of ARCCRABP1 neurons resulted in obesity and a diabetic phenotype in mice. By employing chemogenetic or optogenetic manipulation techniques, the inhibition and activation of ARCCRABP1 neurons resulted in an increase and decrease in food intake, respectively. The axon terminals from these ARCCRABP1 neurons project to several brain regions implicated in feeding regulation such as PVH, BNST, PBN, and NTS. Optogenetic manipulation of these axons within these brain regions resulted in significant alterations of food intake behavior in mice. Furthermore, the electrophysiological studies demonstrated that the activation of ARCCRABP1 neurons induces depolarization in POMC neurons in the hypothalamus. The hormone stimulation studies showed that most of the ARCCRABP1 neurons respond to insulin. Collectively, our findings demonstrate that ARCCRABP1 neurons represent a distinct neuronal subtype involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lihong Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Liling Jin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Yin Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Yang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Jubiao Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Zhenning Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Junxia Qi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Changqing Qu
- Fuyang Normal University, Engineering Technology Research Center of Anti-aging Chinese Herbal Medicine, Fuyang, Anhui, 236037, China
| | - Guanzhong Dong
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China
| | - Yongjie Zhang
- Department of Human Anatomy of Nanjing Medical University, Human Brain Tissue Resource Center of Nanjing Medical University, National Health and Disease Human Brain Tissue Resource Center - sub-center of Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, 210096, China.
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China.
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China.
| |
Collapse
|
13
|
Grajales-Reyes JG. Advances in energy balance & metabolism circuitry. ADVANCES IN GENETICS 2025; 113:1-28. [PMID: 40409794 DOI: 10.1016/bs.adgen.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Advancements in informatics, genetics, and neuroscience have greatly expanded our understanding of how the central nervous system (CNS) regulates energy balance and metabolism. This chapter explores the key neural circuits within the hypothalamus and brainstem that integrate behavioral and physiological processes to maintain metabolic homeostasis. It also examines the dynamic interplay between the CNS and peripheral organs, mediated through hormonal and neuronal signals, which fine-tune appetite, energy expenditure, and body weight. Furthermore, we highlight groundbreaking research that unveils molecular and cellular pathways governing energy regulation, representing a new frontier in addressing obesity and metabolic disorders. Innovative approaches, such as neurogenetic and neuromodulation techniques, are explored as promising strategies for improving weight management and metabolic health. By providing a comprehensive perspective on the mechanisms underlying energy balance, this chapter underscores the transformative potential of emerging therapeutic innovations.
Collapse
Affiliation(s)
- Jose G Grajales-Reyes
- Department of Anesthesiology, Yale University, New Haven, CT, United States; Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
14
|
Vattathara JJ, Diéguez C, López M. Hunger games: a role for cAMP. Am J Physiol Endocrinol Metab 2025; 328:E287-E288. [PMID: 39842455 DOI: 10.1152/ajpendo.00524.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/24/2025]
Affiliation(s)
- Jane Jose Vattathara
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, Spain
| |
Collapse
|
15
|
Luo XD, Tang S, Luo XY, Quzhen L, Xia RH, Wang XW. Mitochondrial regulation of obesity by POMC neurons. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167682. [PMID: 39837429 DOI: 10.1016/j.bbadis.2025.167682] [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: 09/19/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Pro-opiomelanocortin (POMC) neurons, nestled in the hypothalamus, play a pivotal role in the intricate coordination of energy homeostasis and metabolic pathways. These neurons' mitochondria, often hailed as the cell's powerhouses, are crucial for maintaining cellular energy equilibrium and metabolic functionality. Recent research has illuminated the complex interplay between mitochondrial dynamics and POMC neuronal activity, underscoring their critical involvement in the pathogenesis of a spectrum of metabolic disorders, notably obesity and diabetes. This comprehensive review delves into the molecular mechanisms that underlie how mitochondrial function within POMC neurons modulates metabolic regulation. We dissect the impact of mitochondrial dynamics, encompassing fusion, fission, mitophagy, and biogenesis, on the regulation of POMC neuronal activity. Furthermore, we scrutinize the role of mitochondrial dysfunction in POMC neurons in the etiology of obesity, identifying key therapeutic targets within these pathways. We offer an in-depth perspective on the indispensable role of POMC neuronal mitochondria in metabolic regulation and chart future research directions to bridge the existing knowledge gaps in this field.
Collapse
Affiliation(s)
- Xing-Dan Luo
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Si Tang
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Xiang-Yun Luo
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Luosang Quzhen
- The Central Hospital of Qusong County, Shannan, Tibet Autonomous Region 856300, China
| | - Ruo-Han Xia
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China.
| | - Xian-Wang Wang
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Shannan Maternal and Child Health Hospital, Shannan, Xizang 856100, China.
| |
Collapse
|
16
|
Hayashi T, Kumamoto K, Kobayashi T, Hou X, Nagao S, Harada N, Honda S, Shimono Y, Nishio E. Estrogen synthesized in the central nervous system enhances MC4R expression and reduces food intake. FEBS J 2025. [PMID: 39967403 DOI: 10.1111/febs.17426] [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: 06/16/2024] [Revised: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Estrogen is synthesized throughout various tissues in the body, and its production is regulated by the rate-limiting enzyme aromatase (encoded by the Cyp19a1 gene). Notably, aromatase is also expressed in central nervous system cells, allowing for localized estrogen synthesis in regions such as the hypothalamus. Estrogens produced within these neurons are referred to as neuroestrogens. In this study, we investigated the role of neuroestrogens in the regulation of appetite through modulation of hypothalamic pathways in OVX, ArKO, and aromatase-restored mice. Estrogen suppresses appetite by influencing the expression of appetite-regulating peptides, including POMC and NPY, via MC4R. We explored the direct effects of neuroestrogens, independent from ovarian estrogen, on appetite suppression and the underlying molecular mechanisms. We monitored body weight and food intake and evaluated the expression of Cyp19a1, Mc4r, and other appetite-related genes. Our findings indicate that OVX and ArKO mice exhibited increased body weight and food consumption, which correlated with altered expression of Mc4r and Cyp19a1. Conversely, restoration of Cyp19a1 expression in a neuron specific manner significantly decreased food intake and increased Mc4r expression in the hypothalamus. Furthermore, neuroestrogens enhanced leptin responsiveness. Our results imply that neuroestrogens likely contribute to appetite regulation and may be relevant for body weight reduction.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Anatomy and Medical Biology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kanako Kumamoto
- Center for Disease Model and Educational Support, Fujita Health University, Toyoake, Japan
| | - Tatsuya Kobayashi
- Department of Regulatory Science, Research Promotion Unit, Fujita Health University School of Medical Science, Toyoake, Japan
- Reproduction Center, Fujita Health University Haneda Clinic, Otaku, Japan
- Department of Molecular Infectiology, Reproductive Medicine, Chiba University of Graduate School of Medicine, Chiba, Japan
| | - Xinfeng Hou
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shizuko Nagao
- Center for Disease Model and Educational Support, Fujita Health University, Toyoake, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shinichiro Honda
- Department of Biochemistry, Fukuoka School of Pharmaceutical Sciences, Fukuoka University, Japan
| | - Yohei Shimono
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Eiji Nishio
- Department of Obstetrics and Gynecology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
17
|
Wang H, Liu Q, Abouelfetouh MM, Li H, Zhu H, Zhu C, Kiani FA, Ding Y. The role of the hypothalamus-gut microbiota in the pathogenesis of periparturient fatty liver disease in dairy cows. Vet J 2025; 309:106290. [PMID: 39675462 DOI: 10.1016/j.tvjl.2024.106290] [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: 09/20/2024] [Revised: 10/12/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
During the periparturient period, dairy cows experience negative energy balance due to reduced feed intake, leading to adipose tissue breakdown, liver damage, and fat accumulation. This study examined the gut-liver-brain axis to explore the link between fatty liver disease, changes in hypothalamic appetite-related neurons, and microbiome shifts in dairy cows. Thirty cows were monitored, with daily DMI recordings and blood sampling. Postpartum brain, liver, and ileal contents were collected from 10 selected cows, divided into two groups: H-DMI (slight DMI decrease) and L-DMI (severe DMI decrease). The L-DMI group of cows exhibited higher plasma NEFA, BHBA, ALT, and AST levels, along with severe hepatic steatosis and lipid accumulation. Transcriptome sequencing of the hypothalamic arcuate nucleus (ARC) revealed decreased expression of Hypocretin Neuropeptide Precursor (HCRT), orexin-A (OX-A), Orexin Receptor Type 1 (OX1R), and Cannabinoid Receptor 1 (CB1) in the L-DMI group, while Pro-opiomelanocortin (POMC) and Melanocortin 4 Receptor (MC4R) expression increased. Metagenomic analysis of ileal contents showed reduced abundance of Ruminococcus spp. in the L-DMI group, which may be associated with fatty liver disease (FL). Integrated omics analysis showed that increased MC4R expression was correlated with the elevated abundance of bacteria such as Akkermansia glycaniphila, and reduced abundance of species such as Methanobrevubacter thaueri and Ruminococcus spp. Decreased HCRT expression was also linked to Akkermansia glycaniphila. In conclusion, these changes may affect DMI through the OX-A/POMC pathway, with neurological and gut microbiome alterations potentially leading to appetite suppression, negative energy balance, and the development of fatty liver disease.
Collapse
Affiliation(s)
- Haolong Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Liu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Mahmoud M Abouelfetouh
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor 13736, Egypt
| | - Hao Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongmei Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Cong Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Faisal Ayub Kiani
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Department of Clinical Sciences, Faculty of Veterinary Sciences, Bahauddin Zakariya University, Pakistan
| | - Yi Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
18
|
Freire-Agulleiro Ó, Estévez-Salguero Á, Ferreira V, Holleman CL, García-Currás J, González-García I, Nogueiras R, Tena-Sempere M, García-Cáceres C, Diéguez C, López M. SF1-specific deletion of the energy sensor AMPKγ2 induces obesity. Mol Metab 2025; 92:102091. [PMID: 39746605 PMCID: PMC11782900 DOI: 10.1016/j.molmet.2024.102091] [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: 11/29/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE AMP-activated protein kinase (AMPK) is a heterotrimer complex consisting of a catalytic α subunit (α1, α2) with a serine/threonine kinase domain, and two regulatory subunits, β (β1, β2) and γ (γ1, γ2, γ3), encoded by different genes. In the hypothalamus, AMPK plays a crucial role in regulating energy balance, including feeding, energy expenditure, peripheral glucose and lipid metabolism. However, most research on hypothalamic AMPK has concentrated on the catalytic subunits AMPKα1 and AMPKα2, with little focus on the regulatory subunits. METHODS To fill this gap of knowledge, we investigated the effects of selectively deleting the regulatory isoform AMPKγ2, which is a primary "energy sensor", in steroidogenic factor 1 (SF1) neurons of the ventromedial hypothalamic nucleus (VMH). Complete metabolic phenotyping and molecular analyses in brown adipose tissue (BAT), white adipose tissue (WAT) and liver were carried out. RESULTS Our findings reveal that, in contrast to the obesity-protective effect of the genetic deletion of AMPKα subunits, the loss of AMPKγ2 in SF1 neurons leads to a sex-independent and feeding-independent obesity-prone phenotype due to decreased thermogenesis in brown adipose tissue (BAT) and reduced browning of WAT, resulting in lower energy expenditure. Additionally, SF1-Cre AMPKγ2 mice exhibit hepatic lipid accumulation, but surprisingly maintain normal glucose homeostasis. CONCLUSIONS Overall, these results highlight the distinct roles of AMPK subunits within the hypothalamus.
Collapse
Affiliation(s)
- Óscar Freire-Agulleiro
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Ánxela Estévez-Salguero
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Vitor Ferreira
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Cassie Lynn Holleman
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Julia García-Currás
- Biostatech Advice, Training and Innovation in Biostatistics, S.L, Ames, 15895, Spain
| | - Ismael González-García
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Rubén Nogueiras
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba, 14004, Spain
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, 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
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
19
|
Laule C, Rahmouni K. Leptin and Associated Neural Pathways Underlying Obesity-Induced Hypertension. Compr Physiol 2025; 15:e8. [PMID: 40293220 PMCID: PMC12038170 DOI: 10.1002/cph4.8] [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: 10/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 04/30/2025]
Abstract
Obesity rates have surged to pandemic levels, placing tremendous burden on our society. This chronic and complex disease is related to the development of many life-threatening illnesses including cardiovascular diseases. Hypertension caused by obesity increases the risk for cardiovascular mortality and morbidity by promoting stroke, myocardial infarction, congestive heart failure, and end-stage renal disease. Overwhelming evidence supports neural origins for obesity-induced hypertension and pinpoints the adipose-derived hormone, leptin, and the sympathetic nervous system as major causal factors. Hyperleptinemia in obesity is associated with selective leptin resistance where leptin's renal sympathoexcitatory and pressor effects are preserved while the metabolic actions are impaired. Understanding the mechanisms driving this phenomenon is critical for developing effective therapeutics. This review describes the neural mechanisms of obesity-induced hypertension with a focus on the molecular and neuronal substrates of leptin action.
Collapse
Affiliation(s)
- Connor Laule
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| |
Collapse
|
20
|
Yang M, Tian S, Han X, Xu L, You J, Wu M, Cao Y, Jiang Y, Zheng Z, Liu J, Meng F, Li C, Wang X. Interleukin-11Rα2 in the hypothalamic arcuate nucleus affects depression-related behaviors and the AKT-BDNF pathway. Gene 2025; 933:148966. [PMID: 39341516 DOI: 10.1016/j.gene.2024.148966] [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: 01/26/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Depression is a widespread emotional disorder with complex pathogenesis. An essential function of the hypothalamus is to regulate emotional disorders. However, further investigation is required to identify the pathogenic genes and molecular mechanisms that contribute to the onset of depression within the hypothalamus. Through RNA-sequencing analysis, this study identified the upregulated expression of interleukin-11 receptor alpha 2 (IL-11Rα2) in the hypothalamus of mice with chronic unpredictable stress (CUS)-induced depression. This substantial increase in IL-11Rα2, not IL-11Rα1 expression levels in the hypothalamus under the influence of CUS was found to be associated with depression-related behaviors. We further showed that IL-11Rα2 is expressed in the arcuate nucleus (ARC) proopiomelanocortin (POMC) neurons of the hypothalamus. Male and female mice exhibited behaviors association with depression, when IL-11Rα2 or its ligand IL-11 was overexpressed in the ARC POMC neurons through the action of an adeno-associated virus. In addition, reductions in the expression levels of proteins involved in the protein kinase B signaling pathways and brain-derived neurotrophic factor were observed upon overexpression of IL-11Rα2 in the hypothalamic ARC. This study emphasizes the importance of IL-11Rα2 in the hypothalamus ARC in the development of depression, and presents it as a potential novel target for depression treatment.
Collapse
Affiliation(s)
- Mengyu Yang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shulei Tian
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaofeng Han
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lihong Xu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jingjing You
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Min Wu
- Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yifan Cao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuting Jiang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ziteng Zheng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xuezhen Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
21
|
Mahdavi K, Zendehdel M, Zarei H. Decoding the role of ghrelin and its interactions with central signaling pathways in avian appetite regulation. Vet Res Commun 2025; 49:73. [PMID: 39804527 DOI: 10.1007/s11259-025-10644-9] [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: 11/25/2024] [Accepted: 01/05/2025] [Indexed: 03/14/2025]
Abstract
Ghrelin, a peptide hormone primarily produced in the enteroendocrine cells of the gastrointestinal tract, plays a vital role in regulating food intake, and energy balance in avian species. This review examines the complex interactions between ghrelin and the central signaling pathways associated with hunger regulation in birds. In contrast to mammals, where ghrelin typically promotes feeding behavior, its effects in birds appear more nuanced, exhibiting anorexigenic properties under certain conditions. The interactions of ghrelin with central signaling pathways, particularly within the hypothalamus, are explored, highlighting its influence on various neuropeptide systems, including GABAergic, corticotropinergic, opioidergic, dopaminergic, serotonergic, cannabinoidergic, and adrenergic pathways. This article synthesizes current knowledge regarding ghrelin's structure and physiological functions, as well as its interactions with other neuropeptides and hormones that collectively govern avian feeding behaviors. Furthermore, this review proposes future research directions aimed at elucidating the intricate mechanisms underlying appetite control in birds. Insights gained from this analysis may not only enhance our understanding of avian biology and the optimal regulation of their food intake but also inform wildlife management and conservation strategies in response to environmental changes.
Collapse
Affiliation(s)
- Kimia Mahdavi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 14155-6453, Iran
| | - Morteza Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 14155-6453, Iran.
| | - Hamed Zarei
- Department of Biology, Faculty of Basic Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
22
|
Possa‐Paranhos IC, Butts J, Pyszka E, Nelson C, Congdon S, Cho D, Sweeney P. Medial hypothalamic MC3R signalling regulates energy rheostasis in adult mice. J Physiol 2025; 603:379-410. [PMID: 39718394 PMCID: PMC11737543 DOI: 10.1113/jp286699] [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: 04/08/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Although mammals resist both acute weight loss and weight gain, the neural circuitry mediating bi-directional defense against weight change is incompletely understood. Global constitutive deletion of the melanocortin-3-receptor (MC3R) impairs the behavioural response to both anorexic and orexigenic stimuli, with MC3R knockout mice demonstrating increased weight gain following anabolic challenges and increased weight loss following anorexic challenges (i.e. impaired energy rheostasis). However, the brain regions mediating this phenotype are not well understood. Here, we utilized MC3R floxed mice and viral injections of Cre-recombinase to selectively delete MC3R from the medial hypothalamus (MH) in adult mice. Behavioural assays were performed on these animals to test the role of MC3R in MH in the acute response to orexigenic and anorexic challenges. Complementary chemogenetic approaches were used in MC3R-Cre mice to localize and characterize the specific medial hypothalamic brain regions mediating the role of MC3R in energy homeostasis. Finally, we performed RNAscope in situ hybridization to map changes in the mRNA expression of MC3R, pro-opiomelanocortin and agouti-related peptide following energy rheostatic challenges, as well as to characterize the MC3R expressing cells in dorsal MH. Our results demonstrate that MC3R deletion in MH increases feeding and weight gain following high-fat diet feeding, and enhances the anorexic effects of semaglutide, in a sexually dimorphic manner. Furthermore, although the arcuate nucleus exerts an important role in MC3R-mediated effects on energy homeostasis, viral deletion in the dorsal MH also resulted in altered energy rheostasis, indicating that brain regions outside of the arcuate nucleus also contribute to the role of MC3R in energy rheostasis. Together, these results demonstrate that MC3R-mediated effects on energy rheostasis result from the loss of MC3R signalling in medial hypothalamic neurons and suggest an important role for dorsal-MH MC3R signalling in energy rheostasis. KEY POINTS: Melanocortin-3-receptor (MC3R) signalling regulates energy rheostasis in adult mice. Medial hypothalamus regulates energy rheostasis in adult mice. Energy rheostatic stimuli alter mRNA levels of agouti-related peptide, pro-opiomelanocortin and MC3R. Dorsal-medial hypothalamus (DMH) MC3R neurons increase locomotion and energy expenditure. MC3R cell types in DMH are sexually dimorphic.
Collapse
Affiliation(s)
| | - Jared Butts
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| | - Emma Pyszka
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Christina Nelson
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Samuel Congdon
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Dajin Cho
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| | - Patrick Sweeney
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| |
Collapse
|
23
|
Argente J, Farooqi IS, Chowen JA, Kühnen P, López M, Morselli E, Gan HW, Spoudeas HA, Wabitsch M, Tena-Sempere M. Hypothalamic obesity: from basic mechanisms to clinical perspectives. Lancet Diabetes Endocrinol 2025; 13:57-68. [PMID: 39547253 DOI: 10.1016/s2213-8587(24)00283-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 11/17/2024]
Abstract
Despite the diverse nature of obesity, there is compelling genetic, clinical, and experimental evidence that endorses the important contribution of brain circuits to this condition. The hypothalamus contains major regulatory circuits for bodyweight homoeostasis, the deregulation of which can lead to obesity. Although functional perturbation of hypothalamic pathways could lie at the basis of common forms of obesity, the term hypothalamic obesity has been created to define those rare forms of severe obesity where a clear hypothalamic substrate can be identified, either of genetic or acquired origin. An in-depth understanding of the pathogenesis, clinical presentation, and therapeutic targets of hypothalamic obesity relies on the comprehension of the physiological basis of hypothalamic pathways governing bodyweight control, the mechanisms (either genetic or acquired) whereby they are perturbed, and the consequences of such perturbation. In this Review, we provide a synoptic overview of hypothalamic obesity, from basic mechanisms to clinical perspectives, with a major focus on current developments and new avenues for the diagnosis and precise treatment of these rare forms of obesity.
Collapse
Affiliation(s)
- Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain; IMDEA-Food Institute, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.
| | - I Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain; IMDEA-Food Institute, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Peter Kühnen
- Department of Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Centre for Child and Adolescent Health, Berlin, Germany
| | - Miguel López
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Hoong-Wei Gan
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Helen A Spoudeas
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; SUCCESS Charity, London, UK
| | - Martin Wabitsch
- German Centre for Child and Adolescent Health, Berlin, Germany; Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, Ulm, Germany
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.
| |
Collapse
|
24
|
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
|
25
|
Nicolas JC, Lee TH, Quarta C. Can brain neurons change identity? Lessons from obesity. Trends Endocrinol Metab 2024:S1043-2760(24)00297-2. [PMID: 39643545 DOI: 10.1016/j.tem.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024]
Abstract
It has long been thought that the functional identity of mammalian brain neurons is programmed during development and remains stable throughout adult life; however, certain populations of neurons continue to express active regulators of neuronal identity into adulthood. Prolonged exposure to diet-induced metabolic stress induces features of neuronal identity modification in adult mice, and maladaptive changes in neuronal identity maintenance have been linked to cognitive impairment in humans suffering from neurodegenerative diseases often associated with obesity. Here we discuss how, by unraveling the neurological roots of obesity, we may solve the puzzle of whether mammalian brain neurons retain identity plasticity into adulthood, while advancing knowledge of the pathogenic mechanisms at the interface of metabolic and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jean Charles Nicolas
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Thomas H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
26
|
López M, Gualillo O. Rheumatic diseases and metabolism: where centre and periphery meet. Nat Rev Rheumatol 2024; 20:783-794. [PMID: 39478099 DOI: 10.1038/s41584-024-01178-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 11/26/2024]
Abstract
Over the past few decades, the connection between metabolism and various inflammatory and rheumatic diseases has been an area of active investigation. Nonetheless, the precise mechanisms underlying these relationships remain a topic of ongoing debate, owing in part to conflicting data. This discrepancy can be attributed to the predominant focus on peripheral mechanisms in research into the metabolic consequences of rheumatic diseases. However, a wealth of evidence supports the notion that the central nervous system, specifically the hypothalamus, has an important influence on metabolic homeostasis. Notably, links have been established between crucial hypothalamic mechanisms responsible for regulating energy balance (including food intake, thermogenesis, and glucose and lipid metabolism), such as AMP-activated protein kinase, and the pathophysiology of rheumatoid arthritis. This Review aims to comprehensively examine the current understanding of central metabolic control in rheumatic diseases and explore potential therapeutic options that target this pathophysiological mechanism.
Collapse
Affiliation(s)
- Miguel López
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, Spain.
| | - Oreste Gualillo
- Servizo Galego de Saude (SERGAS)-Instituto de Investigación Sanitaria de Santiago (IDIS), the Neuroendocrine Interactions in Rheumatology and Inflammatory Disease (NEIRID) Lab, Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
27
|
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
|
28
|
Lee TH, Nicolas JC, Quarta C. Molecular and functional mapping of the neuroendocrine hypothalamus: a new era begins. J Endocrinol Invest 2024; 47:2627-2648. [PMID: 38878127 DOI: 10.1007/s40618-024-02411-5] [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: 02/23/2024] [Accepted: 06/08/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Recent advances in neuroscience tools for single-cell molecular profiling of brain neurons have revealed an enormous spectrum of neuronal subpopulations within the neuroendocrine hypothalamus, highlighting the remarkable molecular and cellular heterogeneity of this brain area. RATIONALE Neuronal diversity in the hypothalamus reflects the high functional plasticity of this brain area, where multiple neuronal populations flexibly integrate a variety of physiological outputs, including energy balance, stress and fertility, through crosstalk mechanisms with peripheral hormones. Intrinsic functional heterogeneity is also observed within classically 'defined' subpopulations of neuroendocrine neurons, including subtypes with distinct neurochemical signatures, spatial organisation and responsiveness to hormonal cues. AIM The aim of this review is to critically evaluate past and current research on the functional diversity of hypothalamic neuroendocrine neurons and their plasticity. It focuses on how this neuronal plasticity in this brain area relates to metabolic control, feeding regulation and interactions with stress and fertility-related neural circuits. CONCLUSION Our analysis provides an original framework for improving our understanding of the hypothalamic regulation of hormone function and the development of neuroendocrine diseases.
Collapse
Affiliation(s)
- T H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - J-C Nicolas
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - C Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France.
| |
Collapse
|
29
|
Eftychidis V, Ellender TJ, Szymanski J, Minichiello L. Cholecystokinin-expressing neurons of the ventromedial hypothalamic nucleus control energy homeostasis. Front Cell Neurosci 2024; 18:1483368. [PMID: 39529694 PMCID: PMC11550940 DOI: 10.3389/fncel.2024.1483368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
The hypothalamus is the primary center of the brain that regulates energy homeostasis. The ventromedial hypothalamus (VMH) plays a central role in maintaining energy balance by regulating food intake, energy expenditure, and glucose levels. However, the cellular and molecular mechanisms underlying its functions are still poorly understood. Cholecystokinin (CCK) is one of many genes expressed in this hypothalamic nucleus. Peripheral CCK regulates food intake, body weight, and glucose homeostasis. However, current research does not explain the function of CCK neurons in specific nuclei of the hypothalamus and their likely roles in network dynamics related to energy balance and food intake. This study uses genetic and pharmacological methods to examine the role of CCK-expressing neurons in the VMH (CCKVMH). Namely, using a previously generated BAC transgenic line expressing Cre recombinase under the CCK promoter, we performed targeted manipulations of CCKVMH neurons. Histological and transcriptomic database analysis revealed extensive distribution of these neurons in the VMH, with significant heterogeneity in gene expression related to energy balance, including co-expression with PACAP and somatostatin. Pharmacogenetic acute inhibition via Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) resulted in increased food intake and altered meal patterns, characterized by higher meal frequency and shorter intermeal intervals. Furthermore, diphtheria toxin-mediated ablation of CCKVMH neurons led to significant weight gain and hyperphagia over time, increasing meal size and duration. These mice also exhibited impaired glucose tolerance, indicative of disrupted glucose homeostasis. Our findings underscore the integral role of CCKVMH neurons in modulating feeding behavior, energy homeostasis, and glucose regulation. This study enhances our understanding of the neurohormonal mechanisms underlying obesity and metabolic disorders, providing potential targets for therapeutic interventions.
Collapse
Affiliation(s)
| | - Tommas J Ellender
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jacek Szymanski
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
30
|
Ardanaz CG, de la Cruz A, Minhas PS, Hernández-Martín N, Pozo MÁ, Valdecantos MP, Valverde ÁM, Villa-Valverde P, Elizalde-Horcada M, Puerta E, Ramírez MJ, Ortega JE, Urbiola A, Ederra C, Ariz M, Ortiz-de-Solórzano C, Fernández-Irigoyen J, Santamaría E, Karsenty G, Brüning JC, Solas M. Astrocytic GLUT1 reduction paradoxically improves central and peripheral glucose homeostasis. SCIENCE ADVANCES 2024; 10:eadp1115. [PMID: 39423276 PMCID: PMC11488540 DOI: 10.1126/sciadv.adp1115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Astrocytes are considered an essential source of blood-borne glucose or its metabolites to neurons. Nonetheless, the necessity of the main astrocyte glucose transporter, i.e., GLUT1, for brain glucose metabolism has not been defined. Unexpectedly, we found that brain glucose metabolism was paradoxically augmented in mice with astrocytic GLUT1 reduction (GLUT1ΔGFAP mice). These mice also exhibited improved peripheral glucose metabolism especially in obesity, rendering them metabolically healthier. Mechanistically, we observed that GLUT1-deficient astrocytes exhibited increased insulin receptor-dependent ATP release, and that both astrocyte insulin signaling and brain purinergic signaling are essential for improved brain function and systemic glucose metabolism. Collectively, we demonstrate that astrocytic GLUT1 is central to the regulation of brain energetics, yet its depletion triggers a reprogramming of brain metabolism sufficient to sustain energy requirements, peripheral glucose homeostasis, and cognitive function.
Collapse
Affiliation(s)
- Carlos G. Ardanaz
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Aida de la Cruz
- Laboratory of Local Translation in Neurons and Glia, Achucarro Basque Centre for Neuroscience, 48940 Leioa, Spain
| | - Paras S. Minhas
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nira Hernández-Martín
- Unidad de Cartografía Cerebral, Instituto Pluridisciplinar, Universidad Complutense de Madrid, 28040 Madrid, Spain
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Miguel Ángel Pozo
- Unidad de Cartografía Cerebral, Instituto Pluridisciplinar, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Unidad de Cartografía Cerebral, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - M. Pilar Valdecantos
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Department of Metabolism and Cellular Signaling, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid 28029, Spain
- Universidad Francisco de Vitoria, Faculty of Experimental Sciences, Pozuelo de Alarcon, Madrid, Spain
| | - Ángela M. Valverde
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Department of Metabolism and Cellular Signaling, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid 28029, Spain
| | | | | | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Jorge E. Ortega
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Department of Pharmacology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Ainhoa Urbiola
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Cristina Ederra
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Mikel Ariz
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Department of Electrical, Electronic and Communications Engineering, Public University of Navarra, 31006 Pamplona, Spain
| | - Carlos Ortiz-de-Solórzano
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY, USA
| | - Jens C. Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, 50931 Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Maite Solas
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| |
Collapse
|
31
|
Zhang N, Song B, Bai P, Du L, Chen L, Xu Y, Zeng T. Perineuronal nets' role in metabolism. Am J Physiol Endocrinol Metab 2024; 327:E411-E421. [PMID: 39140971 DOI: 10.1152/ajpendo.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Perineuronal nets (PNNs), specialized extracellular matrix (ECM) structures that envelop neurons, have recently been recognized as key players in the regulation of metabolism. This review explores the growing body of knowledge concerning PNNs and their role in metabolic control, drawing insights from recent research and relevant studies. The pivotal role of PNNs in the context of energy balance and whole body blood glucose is examined. This review also highlights novel findings, including the effects of astroglia, microglia, sex and gonadal hormones, nutritional regulation, circadian rhythms, and age on PNNs dynamics. These findings illuminate the complex and multifaceted role of PNNs in metabolic health.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Beite Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li Du
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
32
|
Jovanovic VM, Narisu N, Bonnycastle LL, Tharakan R, Mesch KT, Glover HJ, Yan T, Sinha N, Sen C, Castellano D, Yang S, Blivis D, Ryu S, Bennett DF, Rosales-Soto G, Inman J, Ormanoglu P, LeClair CA, Xia M, Schneider M, Hernandez-Ochoa EO, Erdos MR, Simeonov A, Chen S, Collins FS, Doege CA, Tristan CA. Scalable Hypothalamic Arcuate Neuron Differentiation from Human Pluripotent Stem Cells Suitable for Modeling Metabolic and Reproductive Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601062. [PMID: 39005353 PMCID: PMC11244856 DOI: 10.1101/2024.06.27.601062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The hypothalamus, composed of several nuclei, is essential for maintaining our body's homeostasis. The arcuate nucleus (ARC), located in the mediobasal hypothalamus, contains neuronal populations with eminent roles in energy and glucose homeostasis as well as reproduction. These neuronal populations are of great interest for translational research. To fulfill this promise, we used a robotic cell culture platform to provide a scalable and chemically defined approach for differentiating human pluripotent stem cells (hPSCs) into pro-opiomelanocortin (POMC), somatostatin (SST), tyrosine hydroxylase (TH) and gonadotropin-releasing hormone (GnRH) neuronal subpopulations with an ARC-like signature. This robust approach is reproducible across several distinct hPSC lines and exhibits a stepwise induction of key ventral diencephalon and ARC markers in transcriptomic profiling experiments. This is further corroborated by direct comparison to human fetal hypothalamus, and the enriched expression of genes implicated in obesity and type 2 diabetes (T2D). Genome-wide chromatin accessibility profiling by ATAC-seq identified accessible regulatory regions that can be utilized to predict candidate enhancers related to metabolic disorders and hypothalamic development. In depth molecular, cellular, and functional experiments unveiled the responsiveness of the hPSC-derived hypothalamic neurons to hormonal stimuli, such as insulin, neuropeptides including kisspeptin, and incretin mimetic drugs such as Exendin-4, highlighting their potential utility as physiologically relevant cellular models for disease studies. In addition, differential glucose and insulin treatments uncovered adaptability within the generated ARC neurons in the dynamic regulation of POMC and insulin receptors. In summary, the establishment of this model represents a novel, chemically defined, and scalable platform for manufacturing large numbers of hypothalamic arcuate neurons and serves as a valuable resource for modeling metabolic and reproductive disorders.
Collapse
Affiliation(s)
- Vukasin M. Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Lori L. Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Ravi Tharakan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Kendall T. Mesch
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - Hannah J. Glover
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Hypothalamus Consortium
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Neelam Sinha
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Chaitali Sen
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - David Castellano
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Shu Yang
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Dvir Blivis
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Daniel F. Bennett
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Giovanni Rosales-Soto
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jason Inman
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Christopher A. LeClair
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Martin Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Erick O. Hernandez-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Michael R. Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Shuibing Chen
- Department of Surgery, Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
- Hypothalamus Consortium
| | - Francis S. Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - 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
- Hypothalamus Consortium
| | - Carlos A. Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| |
Collapse
|
33
|
Türküner MS, Yazıcı A, Özcan F. SIK2 Controls the Homeostatic Character of the POMC Secretome Acutely in Response to Pharmacological ER Stress Induction. Cells 2024; 13:1565. [PMID: 39329749 PMCID: PMC11430698 DOI: 10.3390/cells13181565] [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: 08/03/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The neuronal etiology of obesity is centered around a diet-induced inflammatory state in the arcuate nucleus of the hypothalamus, which impairs the functionality of pro-opiomelanocortine neurons (POMCs) responsible for whole-body energy homeostasis and feeding behavior. Intriguingly, systemic salt inducible kinase 2 (SIK2) knockout mice demonstrated reduced food intake and energy expenditure along with modestly dysregulated metabolic parameters, suggesting a causal link between the absence of SIK2 activity in POMCs and the observed phenotype. To test this hypothesis, we conducted a comparative secretomics study from POMC neurons following pharmacologically induced endoplasmic reticulum (ER) stress induction, a hallmark of metabolic inflammation and POMC dysregulation in diet-induced obese (DIO) mice. Our data provide significant in vitro evidence for the POMC-specific SIK2 activity in controlling energy metabolism and feeding in DIO mice by regulating the nature of the related POMC secretome. Our data also suggest that under physiological stress conditions, SIK2 may act as a gatekeeper for the secreted inflammatory factors and signaling molecules critical for cellular survival and energy homeostasis. On the other hand, in the absence of SIK2, the gate opens, leading to a surge of inflammatory cytokines and apoptotic cues concomitant with the dysregulation of POMC neurons.
Collapse
Affiliation(s)
- Mehmet Soner Türküner
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| | - Ayşe Yazıcı
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| | - Ferruh Özcan
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| |
Collapse
|
34
|
Cutugno G, Kyriakidou E, Nadjar A. Rethinking the role of microglia in obesity. Neuropharmacology 2024; 253:109951. [PMID: 38615749 DOI: 10.1016/j.neuropharm.2024.109951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Microglia are the macrophages of the central nervous system (CNS), implying their role in maintaining brain homeostasis. To achieve this, these cells are sensitive to a plethora of endogenous and exogenous signals, such as neuronal activity, cellular debris, hormones, and pathological patterns, among many others. More recent research suggests that microglia are highly responsive to nutrients and dietary variations. In this context, numerous studies have demonstrated their significant role in the development of obesity under calorie surfeit. Because many reviews already exist on this topic, we have chosen to present the state of our reflections on various concepts put forth in the literature, bringing a new perspective whenever possible. Our literature review focuses on studies conducted in the arcuate nucleus of the hypothalamus, a key structure in the control of food intake. Specifically, we present the recent data available on the modifications of microglial energy metabolism following the consumption of an obesogenic diet and their consequences on hypothalamic neuron activity. We also highlight the studies unraveling the mechanisms underlying obesity-related sexual dimorphism. The review concludes with a list of questions that remain to be addressed in the field to achieve a comprehensive understanding of the role of microglia in the regulation of body energy metabolism. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- G Cutugno
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - E Kyriakidou
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - A Nadjar
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
35
|
Gallo R, Teijeiro A, Angulo-Aguado M, Djouder N. IL-17A produced by POMC neurons regulates diet-induced obesity. iScience 2024; 27:110259. [PMID: 39027371 PMCID: PMC11255842 DOI: 10.1016/j.isci.2024.110259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/29/2023] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Overeating leads to obesity, a low-grade inflammatory condition involving interleukin-17A (IL-17A). While pro-opiomelanocortin (POMC) neurons regulate feeding, their connection with IL-17A is not well understood. To impair IL-17A signaling in POMC neurons, IL-17A receptor (Il17ra) was deleted by crossing IL17ra-flox and Pomc-Cre mice. Despite effective deletion, these mice showed no differences in body weight or adiposity compared to control mice, challenging the idea that IL-17A induces obesity through POMC neuron regulation. However, both groups exhibited reduced weight gain and adiposity upon high-fat diet compared to mice carrying only the floxed alleles, suggesting independent effects of Pomc-Cre transgene on body weight. Further analysis reveals that POMC neurons express IL-17A, and reduction in number of POMC neurons in Pomc-Cre mice could be linked to decreased IL-17A expression, which correlates with reduced adipocyte gene expression associated with obesity. Our data underscore an unexpected crosstalk between IL-17A-producing POMC neurons and the endocrine system in obesity regulation.
Collapse
Affiliation(s)
- Rosa Gallo
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Ana Teijeiro
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Mariana Angulo-Aguado
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| |
Collapse
|
36
|
Villa PA, Ruggiero-Ruff RE, Jamieson BB, Campbell RE, Coss D. Obesity Alters POMC and Kisspeptin Neuron Cross Talk Leading to Reduced Luteinizing Hormone in Male Mice. J Neurosci 2024; 44:e0222242024. [PMID: 38744532 PMCID: PMC11236585 DOI: 10.1523/jneurosci.0222-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity is associated with hypogonadism in males, characterized by low testosterone and sperm number. Previous studies determined that these stem from dysregulation of hypothalamic circuitry that regulates reproduction, by unknown mechanisms. Herein, we used mice fed chronic high-fat diet, which mimics human obesity, to determine mechanisms of impairment at the level of the hypothalamus, in particular gonadotropin-releasing hormone (GnRH) neurons that regulate luteinizing hormone (LH), which then regulates testosterone. Consistent with obese humans, we demonstrated lower LH, and lower pulse frequency of LH secretion, but unchanged pituitary responsiveness to GnRH. LH pulse frequency is regulated by pulsatile GnRH secretion, which is controlled by kisspeptin. Peripheral and central kisspeptin injections, and DREADD-mediated activation of kisspeptin neurons, demonstrated that kisspeptin neurons were suppressed in obese mice. Thus, we investigated regulators of kisspeptin secretion. We determined that the LH response to NMDA was lower in obese mice, corresponding to fewer glutamate receptors in kisspeptin neurons, which may be critical for kisspeptin synchronization. Given that kisspeptin neurons also interact with anorexigenic POMC neurons, which are affected by obesity, we examined their cross talk, and determined that the LH response to either DREADD-mediated activation of POMC neurons or central injection of αMSH, a product of POMC, is abolished in obese mice. This was accompanied by diminished levels of αMSH receptor, MC4R, in kisspeptin neurons. Together, our studies determined that obesity leads to the downregulation of receptors that regulate kisspeptin neurons, which is associated with lower LH pulse frequency, leading to lower LH and hypogonadism.
Collapse
Affiliation(s)
- Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Bradley B Jamieson
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| |
Collapse
|
37
|
Heaton EC, Seo EH, Butkovich LM, Yount ST, Gourley SL. Control of goal-directed and inflexible actions by dorsal striatal melanocortin systems, in coordination with the central nucleus of the amygdala. Prog Neurobiol 2024; 238:102629. [PMID: 38763506 PMCID: PMC11198735 DOI: 10.1016/j.pneurobio.2024.102629] [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: 12/19/2023] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
The dorsomedial striatum (DMS) is associated with flexible goal seeking, as opposed to routinized habits. Whether local mechanisms brake this function, for instance when habits may be adaptive, is incompletely understood. We find that a sub-population of dopamine D1 receptor-containing striatal neurons express the melanocortin-4 receptor (MC4R) for α-melanocyte stimulating hormone. These neurons within the DMS are necessary and sufficient for controlling the capacity of mice to flexibly adjust actions based on the likelihood that they will be rewarded. In investigating MC4R function, we found that it suppresses immediate-early gene levels in the DMS and concurrently, flexible goal seeking. MC4R+ neurons receive input from the central nucleus of the amygdala, and behavioral experiments indicate that they are functionally integrated into an amygdalo-striatal circuit that suppresses action flexibility in favor of routine. Publicly available spatial transcriptomics datasets were analyzed for gene transcript correlates of Mc4r expression across the striatal subregions, revealing considerable co-variation in dorsal structures. This insight led to the discovery that the function of MC4R in the dorsolateral striatum complements that in the DMS, in this case suppressing habit-like behavior. Altogether, our findings suggest that striatal MC4R controls the capacity for goal-directed and inflexible actions alike.
Collapse
Affiliation(s)
- Elizabeth C Heaton
- Graduate Program in Neuroscience, Emory University, United States; Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, United States
| | - Esther H Seo
- Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, United States
| | - Laura M Butkovich
- Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, United States
| | - Sophie T Yount
- Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, United States; Graduate Program in Molecular and Systems Pharmacology, Emory University, United States
| | - Shannon L Gourley
- Graduate Program in Neuroscience, Emory University, United States; Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, United States; Graduate Program in Molecular and Systems Pharmacology, Emory University, United States; Children's Healthcare of Atlanta, Atlanta, GA, United States.
| |
Collapse
|
38
|
Lv L, Li Y, Chen X, Qin Z. Transcriptomic analysis reveals the effects of maternal exposure to bisphenol AF on hypothalamic development in male neonatal mice. J Environ Sci (China) 2024; 141:304-313. [PMID: 38408830 DOI: 10.1016/j.jes.2023.05.014] [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: 02/24/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 02/28/2024]
Abstract
Fragmented data suggest that bisphenol AF (BPAF), a chemical widely used in a variety of products, might have potential impacts on the hypothalamus. Here, we employed male neonatal mice following maternal exposure to explore the effects of low-dose BPAF on hypothalamic development by RNA-sequencing. We found that maternal exposure to approximately 50 µg/(kg·day) BPAF from postanal day (PND) 0 to PND 15 altered the hypothalamic transcriptome, primarily involving the pathways and genes associated with extracellular matrix (ECM) and intercellular adhesion, neuroendocrine regulation, and neurological processes. Further RNA analysis confirmed the changes in the expression levels of concerned genes. Importantly, we further revealed that low-dose BPAF posed a stimulatory impact on pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus and induced the browning of inguinal white adipose tissue. All findings indicate that developmental exposure to low-dose BPAF could interfere with hypothalamic development and thereby lead to alterations in the metabolism. Interestingly, 5000 µg/(kg·day) BPAF caused slighter, non-significant or even inverse alterations than the low dose of 50 µg/(kg·day), displaying a dose-independent effect. Further observations suggest that the the dose-independent effects of BPAF might be associated with oxidative stress and inflammatory responses caused by the high dose. Overall, our study highlights a risk of low-dose BPAF to human neuroendocrine regulation and metabolism.
Collapse
Affiliation(s)
- Lin Lv
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanyuan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuanyue Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhanfen Qin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
39
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
40
|
Hao RH, Zhang TP, Jiang F, Liu JH, Dong SS, Li M, Guo Y, Yang TL. Revealing brain cell-stratified causality through dissecting causal variants according to their cell-type-specific effects on gene expression. Nat Commun 2024; 15:4890. [PMID: 38849352 PMCID: PMC11161590 DOI: 10.1038/s41467-024-49263-4] [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: 08/01/2023] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
The human brain has been implicated in the pathogenesis of several complex diseases. Taking advantage of single-cell techniques, genome-wide association studies (GWAS) have taken it a step further and revealed brain cell-type-specific functions for disease loci. However, genetic causal associations inferred by Mendelian randomization (MR) studies usually include all instrumental variables from GWAS, which hampers the understanding of cell-specific causality. Here, we developed an analytical framework, Cell-Stratified MR (csMR), to investigate cell-stratified causality through colocalizing GWAS signals with single-cell eQTL from different brain cells. By applying to obesity-related traits, our results demonstrate the cell-type-specific effects of GWAS variants on gene expression, and indicate the benefits of csMR to identify cell-type-specific causal effect that is often hidden from bulk analyses. We also found csMR valuable to reveal distinct causal pathways between different obesity indicators. These findings suggest the value of our approach to prioritize target cells for extending genetic causation studies.
Collapse
Affiliation(s)
- Ruo-Han Hao
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Tian-Pei Zhang
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Feng Jiang
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Jun-Hui Liu
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Shan-Shan Dong
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Meng Li
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Yan Guo
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China.
| | - Tie-Lin Yang
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China.
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China.
| |
Collapse
|
41
|
Possa-Paranhos IC, Butts J, Pyszka E, Nelson C, Cho D, Sweeney P. Neuroanatomical dissection of the MC3R circuitry regulating energy rheostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590573. [PMID: 38712101 PMCID: PMC11071362 DOI: 10.1101/2024.04.22.590573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Although mammals resist both acute weight loss and weight gain, the neural circuitry mediating bi-directional defense against weight change is incompletely understood. Global constitutive deletion of the melanocortin-3-receptor (MC3R) impairs the behavioral response to both anorexic and orexigenic stimuli, with MC3R knockout mice demonstrating increased weight gain following anabolic challenges and increased weight loss following anorexic challenges (i.e. impaired energy rheostasis). However, the brain regions mediating this phenotype remain incompletely understood. Here, we utilized MC3R floxed mice and viral injections of Cre-recombinase to selectively delete MC3R from medial hypothalamus (MH) in adult mice. Behavioral assays were performed on these animals to test the role of MC3R in MH in the acute response to orexigenic and anorexic challenges. Complementary chemogenetic approaches were used in MC3R-Cre mice to localize and characterize the specific medial hypothalamic brain regions mediating the role of MC3R in energy homeostasis. Finally, we performed RNAscope in situ hybridization to map changes in the mRNA expression of MC3R, POMC, and AgRP following energy rheostatic challenges. Our results demonstrate that MC3R deletion in MH increased feeding and weight gain following acute high fat diet feeding in males, and enhanced the anorexic effects of semaglutide, in a sexually dimorphic manner. Additionally, activation of DMH MC3R neurons increased energy expenditure and locomotion. Together, these results demonstrate that MC3R mediated effects on energy rheostasis result from the loss of MC3R signaling in the medial hypothalamus of adult animals and suggest an important role for DMH MC3R signaling in energy rheostasis.
Collapse
Affiliation(s)
| | - Jared Butts
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| | - Emma Pyszka
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
| | - Christina Nelson
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
| | - Dajin Cho
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| | - Patrick Sweeney
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| |
Collapse
|
42
|
Leon S, Simon V, Lee TH, Steuernagel L, Clark S, Biglari N, Lesté-Lasserre T, Dupuy N, Cannich A, Bellocchio L, Zizzari P, Allard C, Gonzales D, Le Feuvre Y, Lhuillier E, Brochard A, Nicolas JC, Teillon J, Nikolski M, Marsicano G, Fioramonti X, Brüning JC, Cota D, Quarta C. Single cell tracing of Pomc neurons reveals recruitment of 'Ghost' subtypes with atypical identity in a mouse model of obesity. Nat Commun 2024; 15:3443. [PMID: 38658557 PMCID: PMC11043070 DOI: 10.1038/s41467-024-47877-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
The hypothalamus contains a remarkable diversity of neurons that orchestrate behavioural and metabolic outputs in a highly plastic manner. Neuronal diversity is key to enabling hypothalamic functions and, according to the neuroscience dogma, it is predetermined during embryonic life. Here, by combining lineage tracing of hypothalamic pro-opiomelanocortin (Pomc) neurons with single-cell profiling approaches in adult male mice, we uncovered subpopulations of 'Ghost' neurons endowed with atypical molecular and functional identity. Compared to 'classical' Pomc neurons, Ghost neurons exhibit negligible Pomc expression and are 'invisible' to available neuroanatomical approaches and promoter-based reporter mice for studying Pomc biology. Ghost neuron numbers augment in diet-induced obese mice, independent of neurogenesis or cell death, but weight loss can reverse this shift. Our work challenges the notion of fixed, developmentally programmed neuronal identities in the mature hypothalamus and highlight the ability of specialised neurons to reversibly adapt their functional identity to adult-onset obesogenic stimuli.
Collapse
Affiliation(s)
- Stéphane Leon
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Vincent Simon
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Thomas H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Samantha Clark
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Nasim Biglari
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | | | - Nathalie Dupuy
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Astrid Cannich
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Luigi Bellocchio
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Philippe Zizzari
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Delphine Gonzales
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Yves Le Feuvre
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Emeline Lhuillier
- University of Toulouse III Paul Sabatier, INSERM, Institut des Maladies Métaboliques et Cardiovasculaires, U1297, 31400, France; GeT-Santé, Plateforme Génome et Transcriptome, GenoToul, Toulouse, France
| | - Alexandre Brochard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Jean Charles Nicolas
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Jérémie Teillon
- University of Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, F-33000, Bordeaux, France
| | - Macha Nikolski
- University of Bordeaux, Bordeaux Bioinformatics Center, Bordeaux, France
- University of Bordeaux, CNRS, IBGC UMR 5095, Bordeaux, France
| | - Giovanni Marsicano
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Xavier Fioramonti
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
43
|
Qiu S, Wu Q, Wang H, Liu D, Chen C, Zhu Z, Zheng H, Yang G, Li L, Yang M. AZGP1 in POMC neurons modulates energy homeostasis and metabolism through leptin-mediated STAT3 phosphorylation. Nat Commun 2024; 15:3377. [PMID: 38643150 PMCID: PMC11032411 DOI: 10.1038/s41467-024-47684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Zinc-alpha2-glycoprotein (AZGP1) has been implicated in peripheral metabolism; however, its role in regulating energy metabolism in the brain, particularly in POMC neurons, remains unknown. Here, we show that AZGP1 in POMC neurons plays a crucial role in controlling whole-body metabolism. POMC neuron-specific overexpression of Azgp1 under high-fat diet conditions reduces energy intake, raises energy expenditure, elevates peripheral tissue leptin and insulin sensitivity, alleviates liver steatosis, and promotes adipose tissue browning. Conversely, mice with inducible deletion of Azgp1 in POMC neurons exhibit the opposite metabolic phenotypes, showing increased susceptibility to diet-induced obesity. Notably, an increase in AZGP1 signaling in the hypothalamus elevates STAT3 phosphorylation and increases POMC neuron excitability. Mechanistically, AZGP1 enhances leptin-JAK2-STAT3 signaling by interacting with acylglycerol kinase (AGK) to block its ubiquitination degradation. Collectively, these results suggest that AZGP1 plays a crucial role in regulating energy homeostasis and glucose/lipid metabolism by acting on hypothalamic POMC neurons.
Collapse
Affiliation(s)
- Sheng Qiu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Qinan Wu
- Department of Endocrinology, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Hao Wang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Dongfang Liu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Chen Chen
- Endocrinology, SBMS, Faculty of Medicine, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Ling Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Mengliu Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
44
|
Lavoie O, Turmel A, Mattoon P, Desrosiers WJ, Plamondon J, Michael NJ, Caron A. Hypothalamic GABAergic Neurons Expressing Cellular Retinoic Acid Binding Protein 1 (CRABP1) Are Sensitive to Metabolic Status and Liraglutide in Male Mice. Neuroendocrinology 2024; 114:681-697. [PMID: 38631315 PMCID: PMC11232952 DOI: 10.1159/000538716] [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: 11/16/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Owing to their privileged anatomical location, neurons of the arcuate nucleus of the hypothalamus (ARC) play critical roles in sensing and responding to metabolic signals such as leptin and glucagon-like peptide 1 (GLP-1). In addition to the well-known proopiomelanocortin (POMC)- and agouti-related peptide (AgRP)-expressing neurons, subpopulations of GABAergic neurons are emerging as key regulators of energy balance. However, the precise identity of these metabolic neurons is still elusive. Here, we identified and characterized the molecular signature of a novel population of GABAergic neurons of the ARC expressing Cellular retinoic acid binding protein 1 (Crabp1). METHODS Using a combination of immunohistochemistry and in situ hybridization techniques, we investigated the expression of Crabp1 across the mouse brain and characterized the molecular identity of Crabp1ARC neurons. We also determined whether Crabp1ARC neurons are sensitive to fasting, leptin, and GLP1R agonism by assessing cFOS immunoreactivity as a marker of neuronal activity. RESULTS Crabp1ARC neurons represent a novel GABAergic neuronal population robustly enriched in the ARC and are distinct from the prototypical melanocortin neurons. Crabp1ARC neurons overlap with three subpopulations of yet uncharacterized ARC neurons expressing Htr3b, Tbx19, and Tmem215. Notably, Crabp1ARC neurons express receptors for metabolic hormones and their activity is modulated by the nutritional state and GLP1R agonism. CONCLUSION Crabp1ARC neurons represent a novel heterogeneous population of GABAergic neurons sensitive to metabolic status.
Collapse
Affiliation(s)
- Olivier Lavoie
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Audrey Turmel
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Paige Mattoon
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | | | | | - Natalie Jane Michael
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| |
Collapse
|
45
|
Başer Ö, Yavuz Y, Özen DÖ, Özgün HB, Ağuş S, Civaş CC, Atasoy D, Yılmaz B. Effects of chronic high fat diet on mediobasal hypothalamic satiety neuron function in POMC-Cre mice. Mol Metab 2024; 82:101904. [PMID: 38395148 PMCID: PMC10910127 DOI: 10.1016/j.molmet.2024.101904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE The prevalence of obesity has increased over the past three decades. Proopiomelanocortin (POMC) neurons in the hypothalamic arcuate nucleus (ARC) play a vital role in induction of satiety. Chronic consumption of high-fat diet is known to reduce hypothalamic neuronal sensitivity to hormones like leptin, thus contributing to the development and persistence of obesity. The functional and morphological effects of a high-calorie diet on POMC neurons and how these effects contribute to the development and maintenance of the obese phenotype are not fully understood. For this purpose, POMC-Cre transgenic mice model was exposed to high-fat diet (HFD) and at the end of a 3- and 6-month period, electrophysiological and morphological changes, and the role of POMC neurons in homeostatic nutrition and their response to leptin were thoroughly investigated. METHODS Effects of HFD on POMC-satiety neurons in transgenic mice models exposed to chronic high-fat diet were investigated using electrophysiological (patch-clamp), chemogenetic and Cre recombinase advanced technological methods. Leptin, glucose and lipid profiles were determined and analyzed. RESULTS In mice exposed to a high-fat diet for 6 months, no significant changes in POMC dendritic spine number or projection density from POMC neurons to the paraventricular hypothalamus (PVN), lateral hypothalamus (LH), and bed nucleus stria terminalis (BNST) were observed. It was revealed that leptin hormone did not change the electrophysiological activities of POMC neurons in mice fed with HFD for 6 months. In addition, chemogenetic stimulation of POMC neurons increased HFD consumption. In the 3-month HFD-fed group, POMC activation induced an orexigenic response in mice, whereas switching to a standard diet was found to abolish orexigenic behavior in POMC mice. CONCLUSIONS Chronic high fat consumption disrupts the regulation of POMC neuron activation by leptin. Altered POMC neuron activation abolished the neuron's characteristic behavioral anorexigenic response. Change in nutritional content contributes to the reorganization of developing maladaptations.
Collapse
Affiliation(s)
- Özge Başer
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Yavuz Yavuz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Deniz Öykü Özen
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Hüseyin Buğra Özgün
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Sami Ağuş
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Cihan Civan Civaş
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Deniz Atasoy
- University of Iowa, Carver College of Medicine, Department of Neuroscience and Pharmacology, Iowa City, USA
| | - Bayram Yılmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye; Izmir Biomedicine and Genome Center, Izmir, Türkiye.
| |
Collapse
|
46
|
Dittmann MT, Lakatos G, Wainwright JF, Mokrosinski J, Cross E, Farooqi IS, Wallis NJ, Halsey LG, Wilson R, O’Rahilly S, Yeo GS, Raffan E. Low resting metabolic rate and increased hunger due to β-MSH and β-endorphin deletion in a canine model. SCIENCE ADVANCES 2024; 10:eadj3823. [PMID: 38446876 PMCID: PMC10917344 DOI: 10.1126/sciadv.adj3823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
Mutations that perturb leptin-melanocortin signaling are known to cause hyperphagia and obesity, but energy expenditure has not been well studied outside rodents. We report on a common canine mutation in pro-opiomelanocortin (POMC), which prevents production of β-melanocyte-stimulating hormone (β-MSH) and β-endorphin but not α-MSH; humans, similar to dogs, produce α-MSH and β-MSH from the POMC propeptide, but rodents produce only α-MSH. We show that energy expenditure is markedly lower in affected dogs, which also have increased motivational salience in response to a food cue, indicating increased wanting or hunger. There was no difference in satiety at a modified ad libitum meal or in their hedonic response to food, nor disruption of adrenocorticotropic hormone (ACTH) or thyroid axes. In vitro, we show that β-MSH signals comparably to α-MSH at melanocortin receptors. These data implicate β-MSH and β-endorphin as important in determining hunger and moderating energy expenditure and suggest that this role is independent of the presence of α-MSH.
Collapse
Affiliation(s)
- Marie T. Dittmann
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Gabriella Lakatos
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jodie F. Wainwright
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jacek Mokrosinski
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eloise Cross
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I. Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Natalie J. Wallis
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Lewis G. Halsey
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Rory Wilson
- Department of Biosciences, Swansea University, Swansea, UK
| | - Stephen O’Rahilly
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Giles S.H. Yeo
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eleanor Raffan
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Guo H, Xin Y, Wang S, Zhang X, Ren Y, Qiao B, Li H, Wu J, Hao X, Xu L, Yan Y, Zhang H. Hypothalamic POMC neuron-specific knockout of MC4R affects insulin sensitivity by regulating Kir2.1. Mol Med 2024; 30:34. [PMID: 38448811 PMCID: PMC10918880 DOI: 10.1186/s10020-024-00804-z] [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: 12/04/2023] [Accepted: 02/23/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Imbalance in energy regulation is a major cause of insulin resistance and diabetes. Melanocortin-4 receptor (MC4R) signaling at specific sites in the central nervous system has synergistic but non-overlapping functions. However, the mechanism by which MC4R in the arcuate nucleus (ARC) region regulates energy balance and insulin resistance remains unclear. METHODS The MC4Rflox/flox mice with proopiomelanocortin (POMC) -Cre mice were crossed to generate the POMC-MC4Rflox/+ mice. Then POMC-MC4Rflox/+ mice were further mated with MC4Rflox/flox mice to generate the POMC-MC4Rflox/flox mice in which MC4R is selectively deleted in POMC neurons. Bilateral injections of 200 nl of AAV-sh-Kir2.1 (AAV-sh-NC was used as control) were made into the ARC of the hypothalamus. Oxygen consumption, carbon dioxide production, respiratory exchange ratio and energy expenditure were measured by using the CLAMS; Total, visceral and subcutaneous fat was analyzed using micro-CT. Co-immunoprecipitation assays (Co-IP) were used to analyze the interaction between MC4R and Kir2.1 in GT1-7 cells. RESULTS POMC neuron-specific ablation of MC4R in the ARC region promoted food intake, impaired energy expenditure, leading to increased weight gain and impaired systemic glucose homeostasis. Additionally, MC4R ablation reduced the activation of POMC neuron, and is not tissue-specific for peripheral regulation, suggesting the importance of its central regulation. Mechanistically, sequencing analysis and Co-IP assay demonstrated a direct interaction of MC4R with Kir2.1. Knockdown of Kir2.1 in POMC neuron-specific ablation of MC4R restored the effect of MC4R ablation on energy expenditure and systemic glucose homeostasis, indicating by reduced body weight and ameliorated insulin resistance. CONCLUSION Hypothalamic POMC neuron-specific knockout of MC4R affects energy balance and insulin sensitivity by regulating Kir2.1. Kir2.1 represents a new target and pathway that could be targeted in obesity.
Collapse
Affiliation(s)
- Hengru Guo
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Ying Xin
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Saifei Wang
- Department of Endocrinology, Third People's Hospital, Zhengzhou, China
| | - Xiaoning Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yanqi Ren
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Bo Qiao
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Hongjiang Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Wu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao Hao
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Lijun Xu
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yushan Yan
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Haohao Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
48
|
De Solis AJ, Del Río-Martín A, Radermacher J, Chen W, Steuernagel L, Bauder CA, Eggersmann FR, Morgan DA, Cremer AL, Sué M, Germer M, Kukat C, Vollmar S, Backes H, Rahmouni K, Kloppenburg P, Brüning JC. Reciprocal activity of AgRP and POMC neurons governs coordinated control of feeding and metabolism. Nat Metab 2024; 6:473-493. [PMID: 38378998 DOI: 10.1038/s42255-024-00987-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Agouti-related peptide (AgRP)-expressing and proopiomelanocortin (POMC)-expressing neurons reciprocally regulate food intake. Here, we combine non-interacting recombinases to simultaneously express functionally opposing chemogenetic receptors in AgRP and POMC neurons for comparing metabolic responses in male and female mice with simultaneous activation of AgRP and inhibition of POMC neurons with isolated activation of AgRP neurons or isolated inhibition of POMC neurons. We show that food intake is regulated by the additive effect of AgRP neuron activation and POMC neuron inhibition, while systemic insulin sensitivity and gluconeogenesis are differentially modulated by isolated-versus-simultaneous regulation of AgRP and POMC neurons. We identify a neurocircuit engaging Npy1R-expressing neurons in the paraventricular nucleus of the hypothalamus, where activated AgRP neurons and inhibited POMC neurons cooperate to promote food consumption and activate Th+ neurons in the nucleus tractus solitarii. Collectively, these results unveil how food intake is precisely regulated by the simultaneous bidirectional interplay between AgRP and POMC neurocircuits.
Collapse
Affiliation(s)
- Alain J De Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Jan Radermacher
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany
| | - Fynn R Eggersmann
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Anna-Lena Cremer
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Michael Sué
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Maximilian Germer
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Christian Kukat
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Stefan Vollmar
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Heiko Backes
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
49
|
Ramasamy I. Physiological Appetite Regulation and Bariatric Surgery. J Clin Med 2024; 13:1347. [PMID: 38546831 PMCID: PMC10932430 DOI: 10.3390/jcm13051347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Obesity remains a common metabolic disorder and a threat to health as it is associated with numerous complications. Lifestyle modifications and caloric restriction can achieve limited weight loss. Bariatric surgery is an effective way of achieving substantial weight loss as well as glycemic control secondary to weight-related type 2 diabetes mellitus. It has been suggested that an anorexigenic gut hormone response following bariatric surgery contributes to weight loss. Understanding the changes in gut hormones and their contribution to weight loss physiology can lead to new therapeutic treatments for weight loss. Two distinct types of neurons in the arcuate hypothalamic nuclei control food intake: proopiomelanocortin neurons activated by the anorexigenic (satiety) hormones and neurons activated by the orexigenic peptides that release neuropeptide Y and agouti-related peptide (hunger centre). The arcuate nucleus of the hypothalamus integrates hormonal inputs from the gut and adipose tissue (the anorexigenic hormones cholecystokinin, polypeptide YY, glucagon-like peptide-1, oxyntomodulin, leptin, and others) and orexigeneic peptides (ghrelin). Replicating the endocrine response to bariatric surgery through pharmacological mimicry holds promise for medical treatment. Obesity has genetic and environmental factors. New advances in genetic testing have identified both monogenic and polygenic obesity-related genes. Understanding the function of genes contributing to obesity will increase insights into the biology of obesity. This review includes the physiology of appetite control, the influence of genetics on obesity, and the changes that occur following bariatric surgery. This has the potential to lead to the development of more subtle, individualised, treatments for obesity.
Collapse
Affiliation(s)
- Indra Ramasamy
- Department of Blood Sciences, Conquest Hospital, Hastings TN37 7RD, UK
| |
Collapse
|
50
|
Lei Y, Liang X, Sun Y, Yao T, Gong H, Chen Z, Gao Y, Wang H, Wang R, Huang Y, Yang T, Yu M, Liu L, Yi CX, Wu QF, Kong X, Xu X, Liu S, Zhang Z, Liu T. Region-specific transcriptomic responses to obesity and diabetes in macaque hypothalamus. Cell Metab 2024; 36:438-453.e6. [PMID: 38325338 DOI: 10.1016/j.cmet.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/27/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
The hypothalamus plays a crucial role in the progression of obesity and diabetes; however, its structural complexity and cellular heterogeneity impede targeted treatments. Here, we profiled the single-cell and spatial transcriptome of the hypothalamus in obese and sporadic type 2 diabetic macaques, revealing primate-specific distributions of clusters and genes as well as spatial region, cell-type-, and gene-feature-specific changes. The infundibular (INF) and paraventricular nuclei (PVN) are most susceptible to metabolic disruption, with the PVN being more sensitive to diabetes. In the INF, obesity results in reduced synaptic plasticity and energy sensing capability, whereas diabetes involves molecular reprogramming associated with impaired tanycytic barriers, activated microglia, and neuronal inflammatory response. In the PVN, cellular metabolism and neural activity are suppressed in diabetic macaques. Spatial transcriptomic data reveal microglia's preference for the parenchyma over the third ventricle in diabetes. Our findings provide a comprehensive view of molecular changes associated with obesity and diabetes.
Collapse
Affiliation(s)
- Ying Lei
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Xian Liang
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yunong Sun
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Ting Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University School of Medicine, Xi'an, Shanxi 710063, China
| | - Hongyu Gong
- School of Life Sciences, Institues of Biomedical Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanqing Gao
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hui Wang
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yunqi Huang
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Tao Yang
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Miao Yu
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Longqi Liu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xingxing Kong
- School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Xun Xu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China.
| | - Shiping Liu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China.
| | - Zhi Zhang
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China; School of Life Sciences, Institues of Biomedical Sciences, Inner Mongolia University, Hohhot 010000, China.
| |
Collapse
|