1
|
Ariyani W, Yoshikawa C, Tsuneoka H, Amano I, Imayoshi I, Ichinose H, Sumi-Ichinose C, Koibuchi N, Kitamura T, Kohno D. Dopaminergic neurons in the paraventricular hypothalamus extend the food consumption phase. Proc Natl Acad Sci U S A 2025; 122:e2411069122. [PMID: 40153459 PMCID: PMC12002271 DOI: 10.1073/pnas.2411069122] [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: 06/03/2024] [Accepted: 02/25/2025] [Indexed: 03/30/2025] Open
Abstract
Feeding behavior is controlled by various neural networks in the brain that are involved in different feeding phases: Food procurement, consumption, and termination. However, the specific neural circuits controlling the food consumption phase remain poorly understood. Here, we investigated the roles of dopaminergic neurons in the paraventricular nucleus of the hypothalamus (PVH) in the feeding behavior in mice. Our results indicated that the PVH dopaminergic neurons were critical for extending the food consumption phase and involved in the development of obesity through epigenetic mechanisms. These neurons synchronized with proopiomelanocortin neurons during consumption, were stimulated by proopiomelanocortin activation, and projected to the lateral habenula (LHb), where dopamine receptor D2 was involved in the increase in food consumption. In addition, upregulated tyrosine hydroxylase (TH) expression in PVH was associated with obesity and indispensable for obesity induction in mice lacking Dnmt3a. Taken together, our results highlight the roles of PVH dopaminergic neurons in promoting food consumption and obesity induction.
Collapse
Affiliation(s)
- Winda Ariyani
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma371-8512, Japan
| | - Chiharu Yoshikawa
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma371-8512, Japan
| | - Haruka Tsuneoka
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma371-8512, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma371-8511, Japan
| | - Itaru Imayoshi
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto606-8501, Japan
- Department of Brain Development and Regeneration, Graduate School of Biostudies, Kyoto University, Kyoto606-8501, Japan
- Laboratory of Deconstruction of Stem Cells, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Hiroshi Ichinose
- School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Kanagawa226-8501, Japan
| | - Chiho Sumi-Ichinose
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Aichi470-1192, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma371-8511, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma371-8512, Japan
| | - Daisuke Kohno
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma371-8512, Japan
| |
Collapse
|
2
|
Zhang QJ, Luan JC, Gu Q, Song NH, Xia JD. Leptin action on ARC-PVN neural circuit regulates ejaculation behavior by altering sympathetic neuroplasticity. Andrology 2025. [PMID: 39748719 DOI: 10.1111/andr.13833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/08/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Although some studies have revealed the close relationship between leptin and premature ejaculation in clinical practice, whether and how leptin participates in the regulation of ejaculatory behaviors are still unknown. OBJECTIVE To explore the role of leptin on ejaculatory behaviors and its underlying mechanism. MATERIALS AND METHODS Copulation behavior tests were performed after acute and chronic leptin administration at peripheral and central levels. To compare changes in sympathetic nervous system activity, lumbar sympathetic nervous activity, serum noradrenaline levels, and the distribution of sympathetic fibers in vas deferens and seminal vesicles were analyzed. Construction of virus vector, immunohistochemistry, and optogenetics techniques were used to explore the neural circuit mechanism. The density of dendritic spines in parvocellular region of paraventricular nucleus was measured by Golgi staining. RESULTS Acute administration of leptin had no effect on ejaculation behavior in male mice. However, both mount latency and ejaculation latency were significantly shortened, even if serum leptin decreased to normal level, after chronic administration of leptin at peripheral or central level. Additionally, sympathetic fibers in vas deferens and seminal vesicles obviously increased, in which arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus played an important role. Dendritic spine density in parvocellular region increased after chronic leptin administration. DISCUSSION AND CONCLUSION The role of leptin in regulating ejaculation behavior was chronic, not acute, in which leptin chronically modulated sympathetic neuroplasticity via arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus and promoted ejaculatory behaviors. Increased dendritic spine density in parvocellular region of paraventricular nucleus may be involved as well.
Collapse
Affiliation(s)
- Qi-Jie Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Jiao-Chen Luan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Gu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ning-Hong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Dong Xia
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Li YC, Zhang FC, Li D, Weng RX, Yu Y, Gao R, Xu GY. Distinct circuits and molecular targets of the paraventricular hypothalamus decode visceral and somatic pain. Neuron 2024; 112:3734-3749.e5. [PMID: 39326407 DOI: 10.1016/j.neuron.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/17/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
Visceral and somatic pain serve as protective mechanisms against external threats. Accumulated evidence has confirmed that the paraventricular hypothalamus (PVH) plays an important role in the perception of visceral and somatic pain, whereas the exact neural pathways and molecules distinguishing them remain unclear. Here, we report distinct neuronal ensembles within the PVH dedicated to processing visceral and somatic pain signals. An essential discovery is the distinct expression of P2X3R and VIPR2 in visceral and somatic pain-activated PVH neuronal ensembles. Furthermore, visceral pain- and somatic pain-responsive PVH neuronal ensembles project to specific downstream regions, the ventral part of the lateral septal nucleus (LSV) and the caudal part of the zona incerta (ZIC), respectively. These findings unveil that the PVH acts as a pain sorting center that distinctly processes visceral and somatic pain, identifying potential molecular targets for specific pain processing and providing a new framework for comprehending how the brain processes nociceptive information.
Collapse
Affiliation(s)
- Yong-Chang Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China; Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Fu-Chao Zhang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Di Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Rui-Xia Weng
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China; Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, P.R. China
| | - Yang Yu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Rong Gao
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, Jiangsu, P.R. China.
| | - Guang-Yin Xu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China.
| |
Collapse
|
4
|
Xing M, Li Y, Zhang Y, Zhou J, Ma D, Zhang M, Tang M, Ouyang T, Zhang F, Shi X, Sun J, Chen Z, Zhang WJ, Zhang S, Xie X. Paraventricular hypothalamic RUVBL2 neurons suppress appetite by enhancing excitatory synaptic transmission in distinct neurocircuits. Nat Commun 2024; 15:8939. [PMID: 39414808 PMCID: PMC11484884 DOI: 10.1038/s41467-024-53258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The paraventricular hypothalamus (PVH) is crucial for food intake control, yet the presynaptic mechanisms underlying PVH neurons remain unclear. Here, we show that RUVBL2 in the PVH is significantly reduced during energy deficit, and knockout (KO) of PVH RUVBL2 results in hyperphagic obesity in mice. RUVBL2-expressing neurons in the PVH (PVHRUVBL2) exert the anorexigenic effect by projecting to the arcuate hypothalamus, the dorsomedial hypothalamus, and the parabrachial complex. We further demonstrate that PVHRUVBL2 neurons form the synaptic connections with POMC and AgRP neurons in the ARC. PVH RUVBL2 KO impairs the excitatory synaptic transmission by reducing presynaptic boutons and synaptic vesicles near active zone. Finally, RUVBL2 overexpression in the PVH suppresses food intake and protects against diet induced obesity. Together, this study demonstrates an essential role for PVH RUVBL2 in food intake control, and suggests that modulation of synaptic plasticity could be an effective way to curb appetite and obesity.
Collapse
Affiliation(s)
- Mingming Xing
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Juemou Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Danting Ma
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Mengqi Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Minglei Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Ting Ouyang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Fumiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Weiping J Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
5
|
Cai H, Schnapp WI, Mann S, Miscevic M, Shcmit MB, Conteras M, Fang C. Neural circuits regulation of satiation. Appetite 2024; 200:107512. [PMID: 38801994 PMCID: PMC11227400 DOI: 10.1016/j.appet.2024.107512] [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: 02/05/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Terminating a meal after achieving satiation is a critical step in maintaining a healthy energy balance. Despite the extensive collection of information over the last few decades regarding the neural mechanisms controlling overall eating, the mechanism underlying different temporal phases of eating behaviors, especially satiation, remains incompletely understood and is typically embedded in studies that measure the total amount of food intake. In this review, we summarize the neural circuits that detect and integrate satiation signals to suppress appetite, from interoceptive sensory inputs to the final motor outputs. Due to the well-established role of cholecystokinin (CCK) in regulating the satiation, we focus on the neural circuits that are involved in regulating the satiation effect caused by CCK. We also discuss several general principles of how these neural circuits control satiation, as well as the limitations of our current understanding of the circuits function. With the application of new techniques involving sophisticated cell-type-specific manipulation and mapping, as well as real-time recordings, it is now possible to gain a better understanding of the mechanisms specifically underlying satiation.
Collapse
Affiliation(s)
- Haijiang Cai
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Bio 5 Institute and Department of Neurology, University of Arizona, Tucson, AZ, 85721, USA.
| | - Wesley I Schnapp
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Shivani Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Masa Miscevic
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Matthew B Shcmit
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Marco Conteras
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Caohui Fang
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
6
|
Li Y, Kim M, Jiang L, Baron L, Faulkner LD, Olson DP, Li X, Gannot N, Li P, Rui L. SH2B1 Defends Against Energy Imbalance, Obesity, and Metabolic Disease via a Paraventricular Hypothalamus→Dorsal Raphe Nucleus Neurocircuit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400437. [PMID: 38885417 PMCID: PMC11336965 DOI: 10.1002/advs.202400437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/01/2024] [Indexed: 06/20/2024]
Abstract
SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Min‐Hyun Kim
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- College of Health SolutionsArizona State UniversityPhoenixAZ85004USA
| | - Lin Jiang
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Lorelei Baron
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Latrice D. Faulkner
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - David P. Olson
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Xingyu Li
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Noam Gannot
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Peng Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Liangyou Rui
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| |
Collapse
|
7
|
Van Drunen R, Dai Y, Wei H, Fekry B, Noori S, Shivshankar S, Bravo R, Zhao Z, Yoo SH, Justice N, Wu JQ, Tong Q, Eckel-Mahan K. Cell-specific regulation of the circadian clock by BMAL1 in the paraventricular nucleus: Implications for regulation of systemic biological rhythms. Cell Rep 2024; 43:114380. [PMID: 38935503 PMCID: PMC11446153 DOI: 10.1016/j.celrep.2024.114380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/28/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Circadian rhythms are internal biological rhythms driving temporal tissue-specific, metabolic programs. Loss of the circadian transcription factor BMAL1 in the paraventricular nucleus (PVN) of the hypothalamus reveals its importance in metabolic rhythms, but its functions in individual PVN cells are poorly understood. Here, loss of BMAL1 in the PVN results in arrhythmicity of processes controlling energy balance and alters peripheral diurnal gene expression. BMAL1 chromatin immunoprecipitation sequencing (ChIP-seq) and single-nucleus RNA sequencing (snRNA-seq) reveal its temporal regulation of target genes, including oxytocin (OXT), and restoring circulating OXT peaks in BMAL1-PVN knockout (KO) mice rescues absent activity rhythms. While glutamatergic neurons undergo day/night changes in expression of genes involved in cell morphogenesis, astrocytes and oligodendrocytes show gene expression changes in cytoskeletal organization and oxidative phosphorylation. Collectively, our findings show diurnal gene regulation in neuronal and non-neuronal PVN cells and that BMAL1 contributes to diurnal OXT secretion, which is important for systemic diurnal rhythms.
Collapse
Affiliation(s)
- Rachel Van Drunen
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yulin Dai
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haichao Wei
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Baharan Fekry
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sina Noori
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Samay Shivshankar
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Rafael Bravo
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Seung-Hee Yoo
- MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biochemistry and Cell Biology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nicholas Justice
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jia Qian Wu
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qingchun Tong
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kristin Eckel-Mahan
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Ji NN, Cao S, Song XL, Pei B, Jin CY, Fan BF, Jiang H, Xia M. Glutamatergic neurons in the paraventricular nucleus of the hypothalamus participate in the regulation of visceral pain induced by pancreatic cancer in mice. Hepatobiliary Surg Nutr 2024; 13:258-272. [PMID: 38617474 PMCID: PMC11007342 DOI: 10.21037/hbsn-23-442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/22/2023] [Indexed: 04/16/2024]
Abstract
Background Visceral pain induced by pancreatic cancer seriously affects patients' quality of life, and there is no effective treatment, because the mechanism of its neural circuit is unknown. Therefore, the aim of this study is to explore the main neural circuit mechanism regulating visceral pain induced by pancreatic cancer in mice. Methods The mouse model of pancreatic cancer visceral pain was established on C57BL/6N mice by pancreatic injection of mPAKPC-luc cells. Abdominal mechanical hyperalgesia and hunch score were performed to assess visceral pain; the pseudorabies virus (PRV) was used to identify the brain regions innervating the pancreas; the c-fos co-labeling method was used to ascertain the types of activated neurons; in vitro electrophysiological patch-clamp technique was used to record the electrophysiological activity of specific neurons; the calcium imaging technique was used to determine the calcium activity of specific neurons; specific neuron destruction and chemogenetics methods were used to explore whether specific neurons were involved in visceral pain induced by pancreatic cancer. Results The PRV injected into the pancreas was detected in the paraventricular nucleus of the hypothalamus (PVN). Immunofluorescence staining showed that the majority of c-fos were co-labeled with glutamatergic neurons in the PVN. In vitro electrophysiological results showed that the firing frequency of glutamatergic neurons in the PVN was increased. The calcium imaging results showed that the calcium activity of glutamatergic neurons in the PVN was enhanced. Both specific destruction of glutamatergic neurons and chemogenetics inhibition of glutamatergic neurons in the PVN alleviated visceral pain induced by pancreatic cancer. Conclusions Glutamatergic neurons in the PVN participate in the regulation of visceral pain induced by pancreatic cancer in mice, providing new insights for the discovery of effective targets for the treatment of pancreatic cancer visceral pain.
Collapse
Affiliation(s)
- Ning-Ning Ji
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang Cao
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing-Lei Song
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Pei
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Yu Jin
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bi-Fa Fan
- Department of Pain, China-Japan Friendship Hospital, Beijing, China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Xia
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Fernandez G, De Francesco PN, Cornejo MP, Cabral A, Aguggia JP, Duque VJ, Sayar N, Cantel S, Burgos JI, Fehrentz JA, Rorato R, Atasoy D, Mecawi AS, Perello M. Ghrelin Action in the PVH of Male Mice: Accessibility, Neuronal Targets, and CRH Neurons Activation. Endocrinology 2023; 164:bqad154. [PMID: 37823477 PMCID: PMC11491828 DOI: 10.1210/endocr/bqad154] [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: 08/05/2023] [Revised: 09/08/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Julieta P Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Victor J Duque
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Nilufer Sayar
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Juan I Burgos
- Centro de Investigaciones Cardiovasculares “Dr. Horacio Eugenio Cingolani” (CONICET and National University of La Plata), La Plata 1900, Buenos Aires, Argentina
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Rodrigo Rorato
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - André S Mecawi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala 751 05, Sweden
| |
Collapse
|
10
|
Wei R, Li D, Jia S, Chen Y, Wang J. MC4R in Central and Peripheral Systems. Adv Biol (Weinh) 2023; 7:e2300035. [PMID: 37043700 DOI: 10.1002/adbi.202300035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/25/2023] [Indexed: 04/14/2023]
Abstract
Obesity has emerged as a critical and urgent health burden during the current global pandemic. Among multiple genetic causes, melanocortin receptor-4 (MC4R), involved in food intake and energy metabolism regulation through various signaling pathways, has been reported to be the lead genetic factor in severe and early onset obesity and hyperphagia disorders. Most previous studies have illustrated the roles of MC4R signaling in energy intake versus expenditure in the central system, while some evidence indicates that MC4R is also expressed in peripheral systems, such as the gut and endocrine organs. However, its physiopathological function remains poorly defined. This review aims to depict the central and peripheral roles of MC4R in energy metabolism and endocrine hormone homeostasis, the diversity of phenotypes, biased downstream signaling caused by distinct MC4R mutations, and current drug development targeting the receptor.
Collapse
Affiliation(s)
- Ran Wei
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Sheng Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| |
Collapse
|
11
|
Xu Y, Jiang Z, Li H, Cai J, Jiang Y, Otiz-Guzman J, Xu Y, Arenkiel BR, Tong Q. Lateral septum as a melanocortin downstream site in obesity development. Cell Rep 2023; 42:112502. [PMID: 37171957 DOI: 10.1016/j.celrep.2023.112502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/27/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
The melanocortin pathway is well established to be critical for body-weight regulation in both rodents and humans. Despite extensive studies focusing on this pathway, the downstream brain sites that mediate its action are not clear. Here, we found that, among the known paraventricular hypothalamic (PVH) neuron groups, those expressing melanocortin receptors 4 (PVHMc4R) preferably project to the ventral part of the lateral septum (LSv), a brain region known to be involved in emotional behaviors. Photostimulation of PVHMc4R neuron terminals in the LSv reduces feeding and causes aversion, whereas deletion of Mc4Rs or disruption of glutamate release from LSv-projecting PVH neurons causes obesity. In addition, disruption of AMPA receptor function in PVH-projected LSv neurons causes obesity. Importantly, chronic inhibition of PVH- or PVHMc4R-projected LSv neurons causes obesity associated with reduced energy expenditure. Thus, the LSv functions as an important node in mediating melanocortin action on body-weight regulation.
Collapse
Affiliation(s)
- Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hongli Li
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanyan Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joshua Otiz-Guzman
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Liu H, Li X, Li P, Hai R, Li J, Fan Q, Wang X, Chen Y, Cao X, Zhang X, Gao R, Wang K, Du C. Glutamatergic melanocortin-4 receptor neurons regulate body weight. FASEB J 2023; 37:e22920. [PMID: 37078546 DOI: 10.1096/fj.202201786r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/22/2023] [Accepted: 03/30/2023] [Indexed: 04/21/2023]
Abstract
The locus coeruleus (LC), enriched in vesicular glutamate transporter 2 (VGlut2) neurons, is a potential homeostasis-regulating hub. However, the identity of melanocortin-4 receptor (MC4R) neurons in the paraventricular nucleus (PVN) of the hypothalamus, PVNVGlut2::MC4R and LCVGlut2::MC4R regulation of body weight, and axonal projections of LCVGlut2 neurons remain unclear. Conditional knockout of MC4R in chimeric mice was used to confirm the effects of VGlut2. Interscapular brown adipose tissue was injected with pseudorabies virus to study the central nervous system projections. We mapped the LCVGlut2 circuitry. Based on the Cre-LoxP recombination system, specific knockdown of MC4R in VGlut2 neurons resulted in weight gain in chimeric mice. Adeno-associated virus-mediated knockdown of MC4R expression in the PVN and LC had potential superimposed effects on weight gain, demonstrating the importance of VGlut2 neurons. Unlike these wide-ranging efferent projections, the PVN, hypothalamic arcuate nucleus, supraoptic nucleus of the lateral olfactory tegmental nuclei, and nucleus tractus solitarius send excitatory projections to LCVGlut2 neurons. The PVN → LC glutamatergic MC4R long-term neural circuit positively affected weight management and could help treat obesity.
Collapse
Affiliation(s)
- Haodong Liu
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaojing Li
- College of Horticulture and Plant Protection, Inner Mongolia Agricultural University, Hohhot, China
| | - Penghui Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Rihan Hai
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Jiacheng Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Qi Fan
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xing Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Yujie Chen
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Xiaojuan Cao
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Xiaoyu Zhang
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| | - Ruifeng Gao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Kun Wang
- Institute of Cereal and Oil Crops, Hebei Academy of Agricultural and Forestry Sciences, Shijiazhuang, China
- Key Laboratory of Crop Cultivation Physiology and Green Production in Hebei Province, Shijiazhuang, China
| | - Chenguang Du
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Vocational and Technical College, Inner Mongolia Agricultural University, Baotou, China
| |
Collapse
|
13
|
Semple EA, Harberson MT, Xu B, Rashleigh R, Cartwright TL, Braun JJ, Custer AC, Liu C, Hill JW. Melanocortin 4 receptor signaling in Sim1 neurons permits sexual receptivity in female mice. Front Endocrinol (Lausanne) 2023; 14:983670. [PMID: 37033219 PMCID: PMC10080118 DOI: 10.3389/fendo.2023.983670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Female sexual dysfunction affects approximately 40% of women in the United States, yet few therapeutic options exist for these patients. The melanocortin system is a new treatment target for hypoactive sexual desire disorder (HSDD), but the neuronal pathways involved are unclear. Methods In this study, the sexual behavior of female MC4R knockout mice lacking melanocortin 4 receptors (MC4Rs) was examined. The mice were then bred to express MC4Rs exclusively on Sim1 neurons (tbMC4RSim1 mice) or on oxytocin neurons (tbMC4ROxt mice) to examine the effect on sexual responsiveness. Results MC4R knockout mice were found to approach males less and have reduced receptivity to copulation, as indicated by a low lordosis quotient. These changes were independent of body weight. Lordosis behavior was normalized in tbMC4RSim1 mice and improved in tbMC4ROxt mice. In contrast, approach behavior was unchanged in tbMC4RSim1 mice but greatly increased in tbMC4ROxt animals. The changes were independent of melanocortin-driven metabolic effects. Discussion These results implicate MC4R signaling in Oxt neurons in appetitive behaviors and MC4R signaling in Sim1 neurons in female sexual receptivity, while suggesting melanocortin-driven sexual function does not rely on metabolic neural circuits.
Collapse
Affiliation(s)
- Erin A. Semple
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Mitchell T. Harberson
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Baijie Xu
- Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, United States
| | - Rebecca Rashleigh
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Tori L. Cartwright
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Jessica J. Braun
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Amy C. Custer
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Chen Liu
- Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, United States
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, United States
| |
Collapse
|
14
|
Wang F, Chen Y, Lin Y, Wang X, Li K, Han Y, Wu J, Shi X, Zhu Z, Long C, Hu X, Duan S, Gao Z. A parabrachial to hypothalamic pathway mediates defensive behavior. eLife 2023; 12:85450. [PMID: 36930206 PMCID: PMC10023160 DOI: 10.7554/elife.85450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/13/2023] [Indexed: 03/18/2023] Open
Abstract
Defensive behaviors are critical for animal's survival. Both the paraventricular nucleus of the hypothalamus (PVN) and the parabrachial nucleus (PBN) have been shown to be involved in defensive behaviors. However, whether there are direct connections between them to mediate defensive behaviors remains unclear. Here, by retrograde and anterograde tracing, we uncover that cholecystokinin (CCK)-expressing neurons in the lateral PBN (LPBCCK) directly project to the PVN. By in vivo fiber photometry recording, we find that LPBCCK neurons actively respond to various threat stimuli. Selective photoactivation of LPBCCK neurons promotes aversion and defensive behaviors. Conversely, photoinhibition of LPBCCK neurons attenuates rat or looming stimuli-induced flight responses. Optogenetic activation of LPBCCK axon terminals within the PVN or PVN glutamatergic neurons promotes defensive behaviors. Whereas chemogenetic and pharmacological inhibition of local PVN neurons prevent LPBCCK-PVN pathway activation-driven flight responses. These data suggest that LPBCCK neurons recruit downstream PVN neurons to actively engage in flight responses. Our study identifies a previously unrecognized role for the LPBCCK-PVN pathway in controlling defensive behaviors.
Collapse
Affiliation(s)
- Fan Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yuge Chen
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yuxin Lin
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xuze Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Kaiyuan Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yong Han
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Jintao Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xingyi Shi
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Zhenggang Zhu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Chaoying Long
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xiaojun Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
- Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
- The Institute of Brain and Cognitive Sciences, Zhejiang University City CollegeHangzhouChina
- Chuanqi Research and Development Center of Zhejiang UniversityHangzhouChina
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
| |
Collapse
|
15
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
16
|
Li YC, Wang Q, Li MG, Hu SF, Xu GY. A paraventricular hypothalamic nucleus input to ventral of lateral septal nucleus controls chronic visceral pain. Pain 2023; 164:625-637. [PMID: 35994589 PMCID: PMC9916060 DOI: 10.1097/j.pain.0000000000002750] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Irritable bowel syndrome is a functional gastrointestinal disorder characterized by chronic visceral pain with complex etiology and difficult treatment. Accumulated evidence has confirmed that the sensitization of the central nervous system plays an important role in the development of visceral pain, whereas the exact mechanisms of action of the neural pathways remain largely unknown. In this study, a distinct neural circuit was identified from the paraventricular hypothalamic (PVH) to the ventral of lateral septal (LSV) region. This circuit was responsible for regulating visceral pain. In particular, the data indicated that the PVH CaMKIIα-positive neurons inputs to the LSV CaMKIIα-positive neurons were only activated by colorectal distention rather than somatic stimulations. The PVH-LSV CaMKIIα + projection pathway was further confirmed by experiments containing a viral tracer. Optogenetic inhibition of PVH CaMKIIα + inputs to LSV CaMKIIα-positive neurons suppressed visceral pain, whereas selective activation of the PVH-LSV CaMKIIα + projection evoked visceral pain. These findings suggest the critical role of the PVH-LSV CaMKIIα + circuit in regulating visceral pain.
Collapse
Affiliation(s)
- Yong-Chang Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, China
| | - Meng-Ge Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Shu-Fen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
17
|
Wang X, Cui X, Li Y, Li F, Li Y, Dai J, Hu H, Wang X, Sun J, Yang Y, Zhang S. MC4R Deficiency Causes Dysregulation of Postsynaptic Excitatory Synaptic Transmission as a Crucial Culprit for Obesity. Diabetes 2022; 71:2331-2343. [PMID: 35926095 DOI: 10.2337/db22-0162] [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: 02/16/2022] [Accepted: 07/30/2022] [Indexed: 11/13/2022]
Abstract
Melanocortin 4 receptor (MC4R) in the paraventricular nucleus of the hypothalamus (PVH) shows bidirectional characterization in modulating food intake and energy homeostasis. We demonstrate that MC4R knockdown (KD) in the PVH can attenuate AMPA receptor (AMPAR)-mediated postsynaptic responses by altering the phosphorylation of AMPAR GluA1 subunit through the protein kinase A (PKA)-dependent signaling cascade and simultaneously lead to rapid body weight gain. Furthermore, PKA KD in the PVH engendered similar electrophysiological and behavioral phenotypes as in MC4R KD mice. Importantly, we observed that the reduction of AMPAR GluA1 expression not only led to attenuated synaptic responses but also caused body weight gain, suggesting that the aberration of synaptic responses may be one of the crucial pathogeny of obesity. Our study provides the synaptic and molecular explanations of how body weight is regulated by MC4R in the PVH.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Cui
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yang Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yue Li
- Guangwai Community Health Service Center of Xicheng District, Beijing, China
| | - Jinye Dai
- Howard Hughes Medical Institute and Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Han Hu
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Wang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Yan Yang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Abuhattum S, Kotzbeck P, Schlüßler R, Harger A, Ariza de Schellenberger A, Kim K, Escolano JC, Müller T, Braun J, Wabitsch M, Tschöp M, Sack I, Brankatschk M, Guck J, Stemmer K, Taubenberger AV. Adipose cells and tissues soften with lipid accumulation while in diabetes adipose tissue stiffens. Sci Rep 2022; 12:10325. [PMID: 35725987 PMCID: PMC9209483 DOI: 10.1038/s41598-022-13324-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/23/2022] [Indexed: 12/14/2022] Open
Abstract
Adipose tissue expansion involves both differentiation of new precursors and size increase of mature adipocytes. While the two processes are well balanced in healthy tissues, obesity and diabetes type II are associated with abnormally enlarged adipocytes and excess lipid accumulation. Previous studies suggested a link between cell stiffness, volume and stem cell differentiation, although in the context of preadipocytes, there have been contradictory results regarding stiffness changes with differentiation. Thus, we set out to quantitatively monitor adipocyte shape and size changes with differentiation and lipid accumulation. We quantified by optical diffraction tomography that differentiating preadipocytes increased their volumes drastically. Atomic force microscopy (AFM)-indentation and -microrheology revealed that during the early phase of differentiation, human preadipocytes became more compliant and more fluid-like, concomitant with ROCK-mediated F-actin remodelling. Adipocytes that had accumulated large lipid droplets were more compliant, and further promoting lipid accumulation led to an even more compliant phenotype. In line with that, high fat diet-induced obesity was associated with more compliant adipose tissue compared to lean animals, both for drosophila fat bodies and murine gonadal adipose tissue. In contrast, adipose tissue of diabetic mice became significantly stiffer as shown not only by AFM but also magnetic resonance elastography. Altogether, we dissect relative contributions of the cytoskeleton and lipid droplets to cell and tissue mechanical changes across different functional states, such as differentiation, nutritional state and disease. Our work therefore sets the basis for future explorations on how tissue mechanical changes influence the behaviour of mechanosensitive tissue-resident cells in metabolic disorders.
Collapse
Affiliation(s)
- Shada Abuhattum
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum Für Physik Und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Petra Kotzbeck
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt GmbH, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Auenbruggerplatz 2, 8036, Graz, Austria
| | - Raimund Schlüßler
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Alexandra Harger
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt GmbH, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Angela Ariza de Schellenberger
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Kyoohyun Kim
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum Für Physik Und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Joan-Carles Escolano
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum Für Physik Und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Torsten Müller
- JPK Instruments/Bruker, Colditzstr. 34-36, 12099, Berlin, Germany
| | - Jürgen Braun
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Martin Wabitsch
- Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Eythstr. 24, 89075, Ulm, Germany
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt GmbH, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Ingolf Sack
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marko Brankatschk
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum Für Physik Und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt GmbH, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Molecular Cell Biology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Universitätsstrasse 2, 86159, Augsburg, Germany
| | - Anna V Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany.
| |
Collapse
|
19
|
Yuan S, Deng B, Ye Q, Wu Z, Wu J, Wang L, Xu Q, Yao L, Xu N. Excitatory neurons in paraventricular hypothalamus contributed to the mechanism underlying acupuncture regulating the swallowing function. Sci Rep 2022; 12:5797. [PMID: 35388042 PMCID: PMC8987055 DOI: 10.1038/s41598-022-09470-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/11/2022] [Indexed: 11/20/2022] Open
Abstract
Paraventricular hypothalamus (PVH) is demonstrated to regulate stress, feeding behaviors, and other related homeostatic processes. However, no direct evidence has been investigated for the role of PVH in swallowing function. Acupuncture therapy at Lianquan (CV23) acupoint has been reported to improve the swallowing function in clinical trials, but its underlying mechanism still needs to be uncovered. Thus, we aimed to explore whether PVH involved the acupuncture mediated regulating swallowing function. Chemogenetics, electromyography (EMG) recording, and immunofluorescence staining methods were combined to demonstrate that neurons in PVH could be activated by electroacupuncture (EA) stimulation at CV23, and this neuronal cluster was represented as excitatory neurons. Furthermore, we mapped both the inputs and outputs of PVH neurons using viral tracing. The neurons in PVH projected with the brain regions, including parabrachial nucleus (PBN) and the solitary tract nucleus (NTS), which both participated in the swallowing process. The EA function regulating the swallowing was attenuated after inhibiting the neurons in PVH in the post stroke dysphagia. In conclusion, this study suggested that EA at CV23 could regulate swallowing function involving the excitatory neurons in PVH.
Collapse
Affiliation(s)
- Si Yuan
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Bing Deng
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Qiuping Ye
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China.,Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Zhennan Wu
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Junshang Wu
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Lin Wang
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Qin Xu
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Lulu Yao
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China.
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Guangzhou Higher Education Mega Center, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, 232 East Ring Road, Panyu District, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
20
|
Yin L, Zhang L, Luo L, Liu Y, Wang F, Feng Y, Wang H, Han Y, Yan Y, Huang C, Fan S. Berbamine reduces body weight via suppression of small GTPase Rab8a activity and activation of paraventricular hypothalamic neurons in obese mice. Eur J Pharmacol 2022; 916:174679. [PMID: 34982965 DOI: 10.1016/j.ejphar.2021.174679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 11/03/2022]
Abstract
Small GTPase Rab8a is involved in fat-specific protein 27 (Fsp27) mediated lipid droplet accumulation in adipocytes. By screening inhibitors of Rab8a GTPase from a natural compound library, berbamine (BBM), a marketing drug for treatment of leukopenia in China, was identified to inhibit the activity of Rab8a GTPase and block the differentiation of 3T3-L1 adipocytes. Animal study showed that BBM could reduce body weight, improved glucose and lipid metabolic homeostasis in high-fat diet-induced obesity (DIO) C57BL/6 mice and db/db mice. Additional, BBM increased energy expenditure and inhibited food intake in mice but not in lean mice. Moreover, intracerebroventricular injection (i.c.v.) of BBM inhibited feeding behavior and increased c-Fos expression in paraventricular nucleus of the hypothalamus (PVH) of mice. Our data suggest that BBM may improve obesity through the inhibition of Rab8a GTPase activity and the activation of anorexigenic energy-sensing neuron in PVH.
Collapse
Affiliation(s)
- Liufang Yin
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Lijun Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lingling Luo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yalei Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fei Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yaru Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongqing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongli Han
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yingxuan Yan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
21
|
Liu Y, Rao B, Li S, Zheng N, Wang J, Bi L, Xu H. Distinct Hypothalamic Paraventricular Nucleus Inputs to the Cingulate Cortex and Paraventricular Thalamic Nucleus Modulate Anxiety and Arousal. Front Pharmacol 2022; 13:814623. [PMID: 35153786 PMCID: PMC8832877 DOI: 10.3389/fphar.2022.814623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/06/2022] [Indexed: 12/16/2022] Open
Abstract
Insomnia and anxiety are two common clinical diseases that threaten people’s physical and mental health. Insomnia and anxiety may share some similar underlying neural circuit mechanisms in the brain. In this study, we combine techniques including chemo-fMRI, optogenetics, and chemogenetics to reveal that the glutamatergic neurons of the paraventricular hypothalamic nucleus (PVN) regulate both anxiety and arousal through two different downstream neural circuits. Optogenetic activation of the PVN-cingulate cortex (Cg) neural circuit triggers anxiety-like behaviors in mice without affecting the wakefulness, while optogenetic activation of the PVN-paraventricular thalamic nucleus (PVT) neural circuit promotes wakefulness in mice without affecting anxiety-like behaviors. Our research reveals that PVN is a key brain area for controlling anxiety and arousal behaviors. We also provide a neurological explanation for anxiety disorder and insomnia which may offer guidance for treatments including drugs or transcranial magnetic stimulation for the patients.
Collapse
Affiliation(s)
- Ying Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bo Rao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuang Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Ning Zheng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Jie Wang, ; Linlin Bi, ; Haibo Xu,
| | - Linlin Bi
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Wuhan University Center for Pathology and Molecular Diagnostics, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Jie Wang, ; Linlin Bi, ; Haibo Xu,
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Jie Wang, ; Linlin Bi, ; Haibo Xu,
| |
Collapse
|
22
|
Singh U, Jiang J, Saito K, Toth BA, Dickey JE, Rodeghiero SR, Deng Y, Deng G, Xue B, Zhu Z, Zingman LV, Geerling JC, Cui H. Neuroanatomical organization and functional roles of PVN MC4R pathways in physiological and behavioral regulations. Mol Metab 2022; 55:101401. [PMID: 34823066 PMCID: PMC8689242 DOI: 10.1016/j.molmet.2021.101401] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE The paraventricular nucleus of hypothalamus (PVN), an integrative center in the brain, orchestrates a wide range of physiological and behavioral responses. While the PVN melanocortin 4 receptor (MC4R) signaling (PVNMC4R+) is involved in feeding regulation, the neuroanatomical organization of PVNMC4R+ connectivity and its role in other physiological regulations are incompletely understood. Here we aimed to better characterize the input-output organization of PVNMC4R+ neurons and test their physiological functions beyond feeding. METHODS Using a combination of viral tools, we mapped PVNMC4R+ circuits and tested the effects of chemogenetic activation of PVNMC4R+ neurons on thermoregulation, cardiovascular control, and other behavioral responses beyond feeding. RESULTS We found that PVNMC4R+ neurons innervate many different brain regions that are known to be important not only for feeding but also for neuroendocrine and autonomic control of thermoregulation and cardiovascular function, including but not limited to the preoptic area, median eminence, parabrachial nucleus, pre-locus coeruleus, nucleus of solitary tract, ventrolateral medulla, and thoracic spinal cord. Contrary to these broad efferent projections, PVNMC4R+ neurons receive monosynaptic inputs mainly from other hypothalamic nuclei (preoptic area, arcuate and dorsomedial hypothalamic nuclei, supraoptic nucleus, and premammillary nucleus), the circumventricular organs (subfornical organ and vascular organ of lamina terminalis), the bed nucleus of stria terminalis, and the parabrachial nucleus. Consistent with their broad efferent projections, chemogenetic activation of PVNMC4R+ neurons not only suppressed feeding but also led to an apparent increase in heart rate, blood pressure, and brown adipose tissue temperature. These physiological changes accompanied acute transient hyperactivity followed by hypoactivity and resting-like behavior. CONCLUSIONS Our results elucidate the neuroanatomical organization of PVNMC4R+ circuits and shed new light on the roles of PVNMC4R+ pathways in autonomic control of thermoregulation, cardiovascular function, and biphasic behavioral activation.
Collapse
Affiliation(s)
- Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jacob E Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Samuel R Rodeghiero
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, United States
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Leonid V Zingman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Joel C Geerling
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
23
|
Chen CR, Zhong YH, Jiang S, Xu W, Xiao L, Wang Z, Qu WM, Huang ZL. Dysfunctions of the paraventricular hypothalamic nucleus induce hypersomnia in mice. eLife 2021; 10:69909. [PMID: 34787078 PMCID: PMC8631797 DOI: 10.7554/elife.69909] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Hypersomnolence disorder (HD) is characterized by excessive sleep, which is a common sequela following stroke, infection, or tumorigenesis. HD is traditionally thought to be associated with lesions of wake-promoting nuclei. However, lesions of a single wake-promoting nucleus, or even two simultaneously, did not exert serious HD. Therefore, the specific nucleus and neural circuitry for HD remain unknown. Here, we observed that the paraventricular nucleus of the hypothalamus (PVH) exhibited higher c-fos expression during the active period (23:00) than during the inactive period (11:00) in mice. Therefore, we speculated that the PVH, in which most neurons are glutamatergic, may represent one of the key arousal-controlling centers. By using vesicular glutamate transporter 2 (vglut2Cre) mice together with fiber photometry, multichannel electrophysiological recordings, and genetic approaches, we found that PVHvglut2 neurons were most active during wakefulness. Chemogenetic activation of PVHvglut2 neurons induced wakefulness for 9 hr, and photostimulation of PVHvglut2→parabrachial complex/ventral lateral septum circuits immediately drove transitions from sleep to wakefulness. Moreover, lesioning or chemogenetic inhibition of PVHvglut2 neurons dramatically decreased wakefulness. These results indicate that the PVH is critical for arousal promotion and maintenance.
Collapse
Affiliation(s)
- Chang-Rui Chen
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Heng Zhong
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lei Xiao
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zan Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Arcuate and Preoptic Kisspeptin Neurons Exhibit Differential Projections to Hypothalamic Nuclei and Exert Opposite Postsynaptic Effects on Hypothalamic Paraventricular and Dorsomedial Nuclei in the Female Mouse. eNeuro 2021; 8:ENEURO.0093-21.2021. [PMID: 34281980 PMCID: PMC8354717 DOI: 10.1523/eneuro.0093-21.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/21/2021] [Accepted: 07/11/2021] [Indexed: 01/24/2023] Open
Abstract
Kisspeptin (Kiss1) neurons provide indispensable excitatory input to gonadotropin-releasing hormone (GnRH) neurons, which is important for the coordinated release of gonadotropins, estrous cyclicity and ovulation. However, Kiss1 neurons also send projections to many other brain regions within and outside the hypothalamus. Two different populations of Kiss1 neurons, one in the arcuate nucleus (Kiss1ARH) and another in the anteroventral periventricular nucleus (AVPV) and periventricular nucleus (PeN; Kiss1AVPV/PeN) of the hypothalamus are differentially regulated by ovarian steroids, and are believed to form direct contacts with GnRH neurons as well as other neurons. To investigate the projection fields from Kiss1AVPV/PeN and Kiss1ARH neurons in female mice, we used anterograde projection analysis, and channelrhodopsin-assisted circuit mapping (CRACM) to explore their functional input to select target neurons within the paraventricular (PVH) and dorsomedial (DMH) hypothalamus, key preautonomic nuclei. Cre-dependent viral (AAV1-DIO-ChR2 mCherry) vectors were injected into the brain to label the two Kiss1 neuronal populations. Immunocytochemistry (ICC) for mCherry and neuropeptides combined with confocal microscopy was used to determine the projection-fields of both Kiss1 neuronal groups. Whole-cell electrophysiology and optogenetics were used to elucidate the functional input to the PVH and DMH. Our analysis revealed many common but also several clearly separate projection fields between the two different populations of Kiss1 neurons. In addition, optogenetic stimulation of Kiss1 projections to PVH prodynorphin, Vglut2 and DMH CART-expressing neurons, revealed excitatory glutamatergic input from Kiss1ARH neurons and inhibitory GABAergic input from Kiss1AVPV/PeN neurons. Therefore, these steroid-sensitive Kiss1 neuronal groups can differentially control the excitability of target neurons to coordinate autonomic functions with reproduction.
Collapse
|
25
|
Sun EW, Iepsen EW, Pezos N, Lumsden AL, Martin AM, Schober G, Isaacs NJ, Rayner CK, Nguyen NQ, de Fontgalland D, Rabbitt P, Hollington P, Wattchow DA, Hansen T, Holm JC, Liou AP, Jackson VM, Torekov SS, Young RL, Keating DJ. A Gut-Intrinsic Melanocortin Signaling Complex Augments L-Cell Secretion in Humans. Gastroenterology 2021; 161:536-547.e2. [PMID: 33848536 DOI: 10.1053/j.gastro.2021.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Hypothalamic melanocortin 4 receptors (MC4R) are a key regulator of energy homeostasis. Brain-penetrant MC4R agonists have failed, as concentrations required to suppress food intake also increase blood pressure. However, peripherally located MC4R may also mediate metabolic benefits of MC4R activation. Mc4r transcript is enriched in mouse enteroendocrine L cells and peripheral administration of the endogenous MC4R agonist, α-melanocyte stimulating hormone (α-MSH), triggers the release of the anorectic hormones Glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in mice. This study aimed to determine whether pathways linking MC4R and L-cell secretion exist in humans. DESIGN GLP-1 and PYY levels were assessed in body mass index-matched individuals with or without loss-of-function MC4R mutations following an oral glucose tolerance test. Immunohistochemistry was performed on human intestinal sections to characterize the mucosal MC4R system. Static incubations with MC4R agonists were carried out on human intestinal epithelia, GLP-1 and PYY contents of secretion supernatants were assayed. RESULTS Fasting PYY levels and oral glucose-induced GLP-1 secretion were reduced in humans carrying a total loss-of-function MC4R mutation. MC4R was localized to L cells and regulates GLP-1 and PYY secretion from ex vivo human intestine. α-MSH immunoreactivity in the human intestinal epithelia was predominantly localized to L cells. Glucose-sensitive mucosal pro-opiomelanocortin cells provide a local source of α-MSH that is essential for glucose-induced GLP-1 secretion in small intestine. CONCLUSION Our findings describe a previously unidentified signaling nexus in the human gastrointestinal tract involving α-MSH release and MC4R activation on L cells in an autocrine and paracrine fashion. Outcomes from this study have direct implications for targeting mucosal MC4R to treat human metabolic disorders.
Collapse
Affiliation(s)
- Emily W Sun
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Eva W Iepsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Nektaria Pezos
- Nutrition, Diabetes and Metabolism, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, Australia; Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Australia
| | - Amanda L Lumsden
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Alyce M Martin
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Gudrun Schober
- Nutrition, Diabetes and Metabolism, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, Australia; Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Australia
| | - Nichole J Isaacs
- Nutrition, Diabetes and Metabolism, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, Australia; Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Australia
| | - Christopher K Rayner
- Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | - Nam Q Nguyen
- Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | | | - Philippa Rabbitt
- Department of Surgery, Flinders Medical Centre, Bedford Park, Australia
| | - Paul Hollington
- Department of Surgery, Flinders Medical Centre, Bedford Park, Australia
| | - David A Wattchow
- Department of Surgery, Flinders Medical Centre, Bedford Park, Australia
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Christian Holm
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Holbæk University Hospital, Holbæk, Denmark
| | - Alice P Liou
- Cardiovascular and Metabolic Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - V Margaret Jackson
- Cardiovascular and Metabolic Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - Signe S Torekov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | - Richard L Young
- Nutrition, Diabetes and Metabolism, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, Australia; Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Australia.
| | - Damien J Keating
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, Australia.
| |
Collapse
|
26
|
Han Y, Xia G, Srisai D, Meng F, He Y, Ran Y, He Y, Farias M, Hoang G, Tóth I, Dietrich MO, Chen MH, Xu Y, Wu Q. Deciphering an AgRP-serotoninergic neural circuit in distinct control of energy metabolism from feeding. Nat Commun 2021; 12:3525. [PMID: 34112797 PMCID: PMC8192783 DOI: 10.1038/s41467-021-23846-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Contrasting to the established role of the hypothalamic agouti-related protein (AgRP) neurons in feeding regulation, the neural circuit and signaling mechanisms by which they control energy expenditure remains unclear. Here, we report that energy expenditure is regulated by a subgroup of AgRP neurons that send non-collateral projections to neurons within the dorsal lateral part of dorsal raphe nucleus (dlDRN) expressing the melanocortin 4 receptor (MC4R), which in turn innervate nearby serotonergic (5-HT) neurons. Genetic manipulations reveal a bi-directional control of energy expenditure by this circuit without affecting food intake. Fiber photometry and electrophysiological results indicate that the thermo-sensing MC4RdlDRN neurons integrate pre-synaptic AgRP signaling, thereby modulating the post-synaptic serotonergic pathway. Specifically, the MC4RdlDRN signaling elicits profound, bi-directional, regulation of body weight mainly through sympathetic outflow that reprograms mitochondrial bioenergetics within brown and beige fat while feeding remains intact. Together, we suggest that this AgRP neural circuit plays a unique role in persistent control of energy expenditure and body weight, hinting next-generation therapeutic approaches for obesity and metabolic disorders.
Collapse
Affiliation(s)
- Yong Han
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Guobin Xia
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Dollada Srisai
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Fantao Meng
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, United States
| | - Yali Ran
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Monica Farias
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Giang Hoang
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - István Tóth
- Department of Physiology and Biochemistry, Szent Istvan University, Budapeste, Hungary
- Department of Comparative Medicine and Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Marcelo O Dietrich
- Department of Comparative Medicine and Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Miao-Hsueh Chen
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qi Wu
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
27
|
Gonzalez IE, Ramirez-Matias J, Lu C, Pan W, Zhu A, Myers MG, Olson DP. Paraventricular Calcitonin Receptor-Expressing Neurons Modulate Energy Homeostasis in Male Mice. Endocrinology 2021; 162:6218079. [PMID: 33834205 PMCID: PMC8139622 DOI: 10.1210/endocr/bqab072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Indexed: 12/29/2022]
Abstract
The paraventricular nucleus of the hypothalamus (PVH) is a heterogeneous collection of neurons that play important roles in modulating feeding and energy expenditure. Abnormal development or ablation of the PVH results in hyperphagic obesity and defects in energy expenditure whereas selective activation of defined PVH neuronal populations can suppress feeding and may promote energy expenditure. Here, we characterize the contribution of calcitonin receptor-expressing PVH neurons (CalcRPVH) to energy balance control. We used Cre-dependent viral tools delivered stereotaxically to the PVH of CalcR2Acre mice to activate, silence, and trace CalcRPVH neurons and determine their contribution to body weight regulation. Immunohistochemistry of fluorescently-labeled CalcRPVH neurons demonstrates that CalcRPVH neurons are largely distinct from several PVH neuronal populations involved in energy homeostasis; these neurons project to regions of the hindbrain that are implicated in energy balance control, including the nucleus of the solitary tract and the parabrachial nucleus. Acute activation of CalcRPVH neurons suppresses feeding without appreciably augmenting energy expenditure, whereas their silencing leads to obesity that may be due in part due to loss of PVH melanocortin-4 receptor signaling. These data show that CalcRPVH neurons are an essential component of energy balance neurocircuitry and their function is important for body weight maintenance. A thorough understanding of the mechanisms by which CalcRPVH neurons modulate energy balance might identify novel therapeutic targets for the treatment and prevention of obesity.
Collapse
MESH Headings
- Animals
- Eating/physiology
- Energy Metabolism/genetics
- Energy Metabolism/physiology
- Feeding Behavior/physiology
- Homeostasis/physiology
- Hypothalamus/metabolism
- Hypothalamus/physiology
- Male
- Mice
- Mice, Transgenic
- Neurons/metabolism
- Neurons/physiology
- Paraventricular Hypothalamic Nucleus/metabolism
- Paraventricular Hypothalamic Nucleus/physiology
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/physiology
- Receptors, Calcitonin/genetics
- Receptors, Calcitonin/metabolism
- Receptors, Calcitonin/physiology
Collapse
Affiliation(s)
- Ian E Gonzalez
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Division of Endocrinology, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Julliana Ramirez-Matias
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chunxia Lu
- Division of Endocrinology, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Warren Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Allen Zhu
- Division of Endocrinology, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Martin G Myers
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - David P Olson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Division of Endocrinology, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: David P. Olson, MD PhD, Department of Pediatrics, Division of Pediatric Endocrinology, Michigan Medicine, D1205 MPB / SPC 5718, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Fan S, Xu Y, Lu Y, Jiang Z, Li H, Morrill JC, Cai J, Wu Q, Xu Y, Xue M, Arenkiel BR, Huang C, Tong Q. A neural basis for brain leptin action on reducing type 1 diabetic hyperglycemia. Nat Commun 2021; 12:2662. [PMID: 33976218 PMCID: PMC8113586 DOI: 10.1038/s41467-021-22940-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
Central leptin action rescues type 1 diabetic (T1D) hyperglycemia; however, the underlying mechanism and the identity of mediating neurons remain elusive. Here, we show that leptin receptor (LepR)-expressing neurons in arcuate (LepRArc) are selectively activated in T1D. Activation of LepRArc neurons, Arc GABAergic (GABAArc) neurons, or arcuate AgRP neurons, is able to reverse the leptin's rescuing effect. Conversely, inhibition of GABAArc neurons, but not AgRP neurons, produces leptin-mimicking rescuing effects. Further, AgRP neuron function is not required for T1D hyperglycemia or leptin's rescuing effects. Finally, T1D LepRArc neurons show defective nutrient sensing and signs of cellular energy deprivation, which are both restored by leptin, whereas nutrient deprivation reverses the leptin action. Our results identify aberrant activation of LepRArc neurons owing to energy deprivation as the neural basis for T1D hyperglycemia and that leptin action is mediated by inhibiting LepRArc neurons through reversing energy deprivation.
Collapse
Affiliation(s)
- Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yungang Lu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hongli Li
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jessie C Morrill
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- MD Anderson Cancer Center & UT Health Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, USA
| | - Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- MD Anderson Cancer Center & UT Health Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, USA
| | - Qi Wu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Mingshan Xue
- Department of Neuroscience and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
- MD Anderson Cancer Center & UT Health Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX, USA.
- Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
29
|
Vieira WF, Kenzo-Kagawa B, Alvares LE, Cogo JC, Baranauskas V, da Cruz-Höfling MA. Exploring the ability of low-level laser irradiation to reduce myonecrosis and increase Myogenin transcription after Bothrops jararacussu envenomation. Photochem Photobiol Sci 2021; 20:571-583. [PMID: 33895984 DOI: 10.1007/s43630-021-00041-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/08/2021] [Indexed: 01/07/2023]
Abstract
Envenoming caused by snakebites is a very important neglected tropical disease worldwide. The myotoxic phospholipases present in the bothropic venom disrupt the sarcolemma and compromise the mechanisms of energy production, leading to myonecrosis. Photobiomodulation therapy (PBMT) has been used as an effective tool to treat diverse cases of injuries, such as snake venom-induced myonecrosis. Based on that, the aim of this study was to analyze the effects of PBMT through low-level laser irradiation (904 nm) on the muscle regeneration after the myonecrosis induced by Bothrops jararacussu snake venom (Bjssu) injection, focusing on myogenic regulatory factors expression, such as Pax7, MyoD, and Myogenin (MyoG). Male Swiss mice (Mus musculus), 6-8-week-old, weighing 22 ± 3 g were used. Single sub-lethal Bjssu dose or saline was injected into the right mice gastrocnemius muscle. At 3, 24, 48, and 72 h after injections, mice were submitted to PBMT treatment. When finished the periods of 48 and 72 h, mice were euthanized and the right gastrocnemius were collected for analyses. We observed extensive inflammatory infiltrate in all the groups submitted to Bjssu injections. PBMT was able to reduce the myonecrotic area at 48 and 72 h after envenomation. There was a significant increase of MyoG mRNA expression at 72 h after venom injection. The data suggest that beyond the protective effect promoted by PBMT against Bjssu-induced myonecrosis, the low-level laser irradiation was able to stimulate the satellite cells, thus enhancing the muscle repair by improving myogenic differentiation.
Collapse
Affiliation(s)
- Willians Fernando Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato 255, Campinas, SP, 13083-970, Brazil.,Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.,Department of Semiconductors, Instruments and Photonics, Faculty of Electrical Engineering, University of Campinas (UNICAMP), Campinas, SP, Brazil.,Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Bruno Kenzo-Kagawa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Lúcia Elvira Alvares
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - José Carlos Cogo
- Faculty of Biomedical Engineering, Brazil University, Itaquera - São Paulo, SP, Brazil
| | - Vitor Baranauskas
- Department of Semiconductors, Instruments and Photonics, Faculty of Electrical Engineering, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Maria Alice da Cruz-Höfling
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato 255, Campinas, SP, 13083-970, Brazil. .,Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
30
|
Neuroendocrine control of appetite and metabolism. Exp Mol Med 2021; 53:505-516. [PMID: 33837263 PMCID: PMC8102538 DOI: 10.1038/s12276-021-00597-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 02/17/2021] [Indexed: 02/02/2023] Open
Abstract
Body homeostasis is predominantly controlled by hormones secreted by endocrine organs. The central nervous system contains several important endocrine structures, including the hypothalamic-pituitary axis. Conventionally, neurohormones released by the hypothalamus and the pituitary gland (hypophysis) have received much attention owing to the unique functions of the end hormones released by their target peripheral organs (e.g., glucocorticoids released by the adrenal glands). Recent advances in mouse genetics have revealed several important metabolic functions of hypothalamic neurohormone-expressing cells, many of which are not readily explained by the action of the corresponding classical downstream hormones. Notably, the newly identified functions are better explained by the action of conventional neurotransmitters (e.g., glutamate and GABA) that constitute a neuronal circuit. In this review, we discuss the regulation of appetite and metabolism by hypothalamic neurohormone-expressing cells, with a focus on the distinct contributions of neurohormones and neurotransmitters released by these neurons.
Collapse
|
31
|
Houtz J, Liao GY, An JJ, Xu B. Discrete TrkB-expressing neurons of the dorsomedial hypothalamus regulate feeding and thermogenesis. Proc Natl Acad Sci U S A 2021; 118:e2017218118. [PMID: 33468645 PMCID: PMC7848633 DOI: 10.1073/pnas.2017218118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mutations in the TrkB neurotrophin receptor lead to profound obesity in humans, and expression of TrkB in the dorsomedial hypothalamus (DMH) is critical for maintaining energy homeostasis. However, the functional implications of TrkB-fexpressing neurons in the DMH (DMHTrkB) on energy expenditure are unclear. Additionally, the neurocircuitry underlying the effect of DMHTrkB neurons on energy homeostasis has not been explored. In this study, we show that activation of DMHTrkB neurons leads to a robust increase in adaptive thermogenesis and energy expenditure without altering heart rate or blood pressure, while silencing DMHTrkB neurons impairs thermogenesis. Furthermore, we reveal neuroanatomically and functionally distinct populations of DMHTrkB neurons that regulate food intake or thermogenesis. Activation of DMHTrkB neurons projecting to the raphe pallidus (RPa) stimulates thermogenesis and increased energy expenditure, whereas DMHTrkB neurons that send collaterals to the paraventricular hypothalamus (PVH) and preoptic area (POA) inhibit feeding. Together, our findings provide evidence that DMHTrkB neuronal activity plays an important role in regulating energy expenditure and delineate distinct neurocircuits that underly the separate effects of DMHTrkB neuronal activity on food intake and thermogenesis.
Collapse
Affiliation(s)
- Jessica Houtz
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | - Guey-Ying Liao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | - Juan Ji An
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| |
Collapse
|
32
|
Kwon E, Joung HY, Liu SM, Chua SC, Schwartz GJ, Jo YH. Optogenetic stimulation of the liver-projecting melanocortinergic pathway promotes hepatic glucose production. Nat Commun 2020; 11:6295. [PMID: 33293550 PMCID: PMC7722761 DOI: 10.1038/s41467-020-20160-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 11/18/2020] [Indexed: 12/18/2022] Open
Abstract
The central melanocortin system plays a fundamental role in the control of feeding and body weight. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) also regulate overall glucose homeostasis via insulin-dependent and -independent pathways. Here, we report that a subset of ARC POMC neurons innervate the liver via preganglionic parasympathetic acetylcholine (ACh) neurons in the dorsal motor nucleus of the vagus (DMV). Optogenetic stimulation of this liver-projecting melanocortinergic pathway elevates blood glucose levels that is associated with increased expression of hepatic gluconeogenic enzymes in female and male mice. Pharmacological blockade and knockdown of the melanocortin-4 receptor gene in the DMV abolish this stimulation-induced effect. Activation of melanocortin-4 receptors inhibits DMV cholinergic neurons and optogenetic inhibition of liver-projecting parasympathetic cholinergic fibers increases blood glucose levels. This elevated blood glucose is not due to altered pancreatic hormone release. Interestingly, insulin-induced hypoglycemia increases ARC POMC neuron activity. Hence, this liver-projecting melanocortinergic circuit that we identified may play a critical role in the counterregulatory response to hypoglycemia.
Collapse
Affiliation(s)
- Eunjin Kwon
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hye-Young Joung
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shun-Mei Liu
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Streamson C Chua
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gary J Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Young-Hwan Jo
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA.
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
33
|
Berruien NNA, Smith CL. Emerging roles of melanocortin receptor accessory proteins (MRAP and MRAP2) in physiology and pathophysiology. Gene 2020; 757:144949. [PMID: 32679290 DOI: 10.1016/j.gene.2020.144949] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 01/07/2023]
Abstract
Melanocortin-2 receptor accessory protein (MRAP) has an unusual dual topology and influences the expression, localisation, signalling and internalisation of the melanocortin receptor 2 (MC2); the adrenocorticotropic hormone (ACTH) receptor. Mutations in MRAP are associated with familial glucocorticoid deficiency type-2 and evidence is emerging of the importance of MRAP in adrenal development and ACTH signalling. Human MRAP has two functional splice variants: MRAP-α and MRAP-β, unlike MRAP-β, MRAP-α has little expression in brain but is highly expressed in ovary. MRAP2, identified through whole human genome sequence analysis, has approximately 40% sequence homology to MRAP. MRAP2 facilitates MC2 localisation to the cell surface but not ACTH signalling. MRAP and MRAP2 have been found to regulate the surface expression and signalling of all melanocortin receptors (MC1-5). Additionally, MRAP2 moderates the signalling of the G-protein coupled receptors (GCPRs): orexin, prokineticin and GHSR1a; the ghrelin receptor. Whilst MRAP appears to be mainly involved in glucocorticoid synthesis, an important role is emerging for MRAP2 in regulating appetite and energy homeostasis. Transgenic models indicate the importance of MRAP in adrenal gland formation. Like MC3R and MC4R knockout mice, MRAP2 knockout mice have an obese phenotype. In vitro studies indicate that MRAP2 enhances the MC3 and MC4 response to the agonist αMSH, which, like ACTH, is produced through precursor polypeptide proopiomelanocortin (POMC) cleavage. Analysis of cohorts of individuals with obesity have revealed several MRAP2 genetic variants with loss of function mutations which are causative of monogenic hyperphagic obesity with hyperglycaemia and hypertension. MRAP2 may also be associated with female infertility. This review summarises current knowledge of MRAP and MRAP2, their influence on GPCR signalling, and focusses on pathophysiology, particularly familial glucocorticoid deficiency type-2 and obesity.
Collapse
Affiliation(s)
- Nasrin N A Berruien
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK.
| | - Caroline L Smith
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK.
| |
Collapse
|
34
|
Navarro VM. Metabolic regulation of kisspeptin - the link between energy balance and reproduction. Nat Rev Endocrinol 2020; 16:407-420. [PMID: 32427949 PMCID: PMC8852368 DOI: 10.1038/s41574-020-0363-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2020] [Indexed: 12/17/2022]
Abstract
Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.
Collapse
Affiliation(s)
- Víctor M Navarro
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Graduate Program in Neuroscience, Boston, MA, USA.
| |
Collapse
|
35
|
Kim ER, Xu Y, Cassidy RM, Lu Y, Yang Y, Tian J, Li DP, Van Drunen R, Ribas-Latre A, Cai ZL, Xue M, Arenkiel BR, Eckel-Mahan K, Xu Y, Tong Q. Paraventricular hypothalamus mediates diurnal rhythm of metabolism. Nat Commun 2020; 11:3794. [PMID: 32732906 PMCID: PMC7393104 DOI: 10.1038/s41467-020-17578-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 07/09/2020] [Indexed: 12/18/2022] Open
Abstract
Defective rhythmic metabolism is associated with high-fat high-caloric diet (HFD) feeding, ageing and obesity; however, the neural basis underlying HFD effects on diurnal metabolism remains elusive. Here we show that deletion of BMAL1, a core clock gene, in paraventricular hypothalamic (PVH) neurons reduces diurnal rhythmicity in metabolism, causes obesity and diminishes PVH neuron activation in response to fast-refeeding. Animal models mimicking deficiency in PVH neuron responsiveness, achieved through clamping PVH neuron activity at high or low levels, both show obesity and reduced diurnal rhythmicity in metabolism. Interestingly, the PVH exhibits BMAL1-controlled rhythmic expression of GABA-A receptor γ2 subunit, and dampening rhythmicity of GABAergic input to the PVH reduces diurnal rhythmicity in metabolism and causes obesity. Finally, BMAL1 deletion blunts PVH neuron responses to external stressors, an effect mimicked by HFD feeding. Thus, BMAL1-driven PVH neuron responsiveness in dynamic activity changes involving rhythmic GABAergic neurotransmission mediates diurnal rhythmicity in metabolism and is implicated in diet-induced obesity.
Collapse
Affiliation(s)
- Eun Ran Kim
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Yuanzhong Xu
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Ryan M Cassidy
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
- Graduate Program in Neuroscience of MD Anderson and UTHealth Graduate School, Houston, TX, 77030, USA
| | - Yungang Lu
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jinbin Tian
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
- Department of Integrative Physiology and Pharmacology, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - De-Pei Li
- Department of Critical Care and Respiratory Care, Division of Anesthesiology, Critical Care and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rachel Van Drunen
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
- Graduate Program in Neuroscience of MD Anderson and UTHealth Graduate School, Houston, TX, 77030, USA
| | - Aleix Ribas-Latre
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Zhao-Lin Cai
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Benjamin R Arenkiel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Kristin Eckel-Mahan
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA.
- Graduate Program in Neuroscience of MD Anderson and UTHealth Graduate School, Houston, TX, 77030, USA.
- Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
36
|
Link R, Veiksina S, Tahk MJ, Laasfeld T, Paiste P, Kopanchuk S, Rinken A. The constitutive activity of melanocortin-4 receptors in cAMP pathway is allosterically modulated by zinc and copper ions. J Neurochem 2019; 153:346-361. [PMID: 31792980 DOI: 10.1111/jnc.14933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/06/2019] [Accepted: 11/29/2019] [Indexed: 01/25/2023]
Abstract
Melanocortin-4 receptors (MC4 R) are unique among G-protein-coupled receptors (GPCRs) as they have endogenous ligands that can exhibit inverse agonistic properties in the case of elevated basal activity. It is known that the constitutive activity of GPCRs strongly affects the ligand-dependent physiological responses, but little is known about these regulatory mechanisms. Since several metal ions have been shown to be important modulators of the signal transduction of GPCRs, we hypothesized that metal ions regulate the basal activity of MC4 Rs. Implementation of a fluorescence anisotropy assay and novel redshifted fluorescent peptides enabled kinetic characterization of ligand binding to MC4 R expressed on budded baculoviruses. We show that Ca2+ is required for high-affinity ligand binding, but Zn2+ and Cu2+ in the presence of Ca2+ behave as negative allosteric modulators of ligand binding to MC4 R. FRET-based cAMP biosensor was used to measure the activation of MC4 R stably expressed in CHO-K1 cells. At low micromolar concentrations, Zn2+ caused MC4 R-dependent activation of the cAMP pathway, whereas Cu2+ reduced the activity of MC4 R even below the basal level. These findings indicate that at physiologically relevant concentrations can Zn2+ and Cu2+ function as MC4 R agonists or inverse agonists, respectively. This means that depending on the level of constitutive activity induced by Zn2+ ions, the pharmacological effect of orthosteric ligands of MC4 R can be switched from a partial to an inverse agonist. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Science badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Reet Link
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Santa Veiksina
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Päärn Paiste
- Department of Geology, University of Tartu, Tartu, Estonia
| | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| |
Collapse
|
37
|
AY O, OI O, FO Y, AM A, IO A, OJ O. Oral Monosodium Glutamate Differentially Affects Open-Field Behaviours, Behavioural Despair and Place Preference in Male and Female Mice. ACTA ACUST UNITED AC 2019. [DOI: 10.2174/2211556008666181213160527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background:
Monosodium glutamate (MSG) is a flavour enhancer which induces
behavioural changes in animals. However the influence of sex on the behavioural response
to MSG has not been investigated.
Objective:
The sex-differential effects of MSG on open-field behaviours, anxiety-related
behaviour, behavioural despair, place-preference, and plasma/brain glutamate levels in
adult mice were assessed.
Methods:
Mice were assigned to three groups (1-3), based on the models used to assess
behaviours. Animals in group 1 were for the elevated-plus maze and tail-suspension paradigms,
group 2 for the open-field and forced-swim paradigms, while mice in group 3 were
for observation in the conditioned place preference paradigm. Mice in all groups were further
assigned into five subgroups (10 males and 10 females), and administered vehicle (distilled
water at 10 ml/kg) or one of four doses of MSG (20, 40, 80 and 160 mg/kg) daily for
6 weeks, following which they were exposed to the behavioural paradigms. At the end of
the behavioural tests, the animals were sacrificed, and blood was taken for estimation of
glutamate levels. The brains were also homogenised for estimation of glutamate levels.
Results:
MSG was associated with a reduction in locomotion in males and females (except
at 160 mg/kg, male), an anxiolytic response in females, an anxiogenic response in males,
and decreased behavioural despair in both sexes (females more responsive). Postconditioning
MSG-associated place-preference was significantly higher in females. Plasma/
brain glutamate was not significantly different between sexes.
Conclusion:
Repeated MSG administration alters a range of behaviours in a sex-dependent
manner in mice.
Collapse
Affiliation(s)
- Onaolapo AY
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Olawore OI
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Yusuf FO
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adeyemo AM
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adewole IO
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Onaolapo OJ
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| |
Collapse
|
38
|
Semple E, Shalabi F, Hill JW. Oxytocin Neurons Enable Melanocortin Regulation of Male Sexual Function in Mice. Mol Neurobiol 2019; 56:6310-6323. [PMID: 30756300 PMCID: PMC6684847 DOI: 10.1007/s12035-019-1514-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
The melanocortin pathway has been implicated in both metabolism and sexual function. When the melanocortin 4 receptor (MC4R) is knocked out globally, male mice display obesity, low sexual desire, and copulatory difficulties; however, it is unclear whether these phenotypes are interdependent. To elucidate the neuronal circuitry involved in sexual dysfunction in MC4R knockouts, we re-expressed the MC4R in these mice exclusively on Sim1 neurons (tbMC4RSim1 mice) or on a subset of Sim1 neurons, namely oxytocin neurons (tbMC4Roxt mice). The groups were matched at young ages to control for the effects of obesity. Interestingly, young MC4R null mice had no deficits in sexual motivation or erectile function. However, MC4R null mice were found to have an increased latency to reach ejaculation compared to control mice, which was restored in both tbMC4RSim1 and tbMC4Roxt mice. These results indicate that melanocortin signaling via the MC4R on oxytocin neurons is important for normal ejaculation independent of the male's metabolic health.
Collapse
Affiliation(s)
- Erin Semple
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Firas Shalabi
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, USA.
| |
Collapse
|
39
|
mTORC1 and CB1 receptor signaling regulate excitatory glutamatergic inputs onto the hypothalamic paraventricular nucleus in response to energy availability. Mol Metab 2019; 28:151-159. [PMID: 31420305 PMCID: PMC6822143 DOI: 10.1016/j.molmet.2019.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE The hypothalamic paraventricular nucleus (PVN) is a key target of the melanocortin system, which orchestrates behavioral and metabolic responses depending on energy availability. The mechanistic target of rapamycin complex 1 (mTORC1) and the endocannabinoid type 1 receptor (CB1R) pathways are two key signaling systems involved in the regulation of energy balance whose activity closely depends upon energy availability. Here we tested the hypothesis that modulation of mTORC1 and CB1R signaling regulates excitatory glutamatergic inputs onto the PVN. METHODS Patch-clamp recordings in C57BL/6J mice, in mice lacking the mTORC1 component Rptor or CB1R in pro-opio-melanocortin (POMC) neurons, combined with pharmacology targeting mTORC1, the melanocortin receptor type 4 (MC4R), or the endocannabinoid system under chow or a hypercaloric diet. RESULTS Acute pharmacological inhibition of mTORC1 in C57BL/6J mice decreased glutamatergic inputs onto the PVN via a mechanism requiring modulation of MC4R, endocannabinoid 2-AG mobilization by PVN parvocellular neurons, and retrograde activation of presynaptic CB1R. Further electrophysiology studies using mice lacking mTORC1 activity or CB1R in POMC neurons indicated that the observed effects involved mTORC1 and CB1R-dependent regulation of glutamate release from POMC neurons. Finally, energy surfeit caused by hypercaloric high-fat diet feeding, rapidly and time-dependently altered the glutamatergic inputs onto parvocellular neurons and the ability of mTORC1 and CB1R signaling to modulate such excitatory activity. CONCLUSIONS These findings pinpoint the relationship between mTORC1 and endocannabinoid-CB1R signaling in the regulation of the POMC-mediated glutamatergic inputs onto PVN parvocellular neurons and its rapid alteration in conditions favoring the development of obesity.
Collapse
|
40
|
Xu Y, Lu Y, Cassidy RM, Mangieri LR, Zhu C, Huang X, Jiang Z, Justice NJ, Xu Y, Arenkiel BR, Tong Q. Identification of a neurocircuit underlying regulation of feeding by stress-related emotional responses. Nat Commun 2019; 10:3446. [PMID: 31371721 PMCID: PMC6671997 DOI: 10.1038/s41467-019-11399-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/13/2019] [Indexed: 12/12/2022] Open
Abstract
Feeding is known to be profoundly affected by stress-related emotional states and eating disorders are comorbid with psychiatric symptoms and altered emotional responses. The neural basis underlying feeding regulation by stress-related emotional changes is poorly understood. Here, we identify a novel projection from the paraventricular hypothalamus (PVH) to the ventral lateral septum (LSv) that shows a scalable regulation on feeding and behavioral changes related to emotion. Weak photostimulation of glutamatergic PVH→LSv terminals elicits stress-related self-grooming and strong photostimulation causes fear-related escape jumping associated with respective weak and strong inhibition on feeding. In contrast, inhibition of glutamatergic inputs to LSv increases feeding with signs of reduced anxiety. LSv-projecting neurons are concentrated in rostral PVH. LSv and LSv-projecting PVH neurons are activated by stressors in vivo, whereas feeding bouts were associated with reduced activity of these neurons. Thus, PVH→LSv neurotransmission underlies dynamic feeding by orchestrating emotional states, providing a novel neural circuit substrate underlying comorbidity between eating abnormalities and psychiatric disorders.
Collapse
Affiliation(s)
- Yuanzhong Xu
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Yungang Lu
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Ryan M Cassidy
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA.,Graduate Program in Neuroscience of the University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Leandra R Mangieri
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA.,Graduate Program in Neuroscience of the University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Canjun Zhu
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Xugen Huang
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Zhiying Jiang
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Nicholas J Justice
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA.,Graduate Program in Neuroscience of the University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neuroscience and Jan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, 77030, USA. .,Graduate Program in Neuroscience of the University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA. .,Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Mangieri LR, Jiang Z, Lu Y, Xu Y, Cassidy RM, Justice N, Xu Y, Arenkiel BR, Tong Q. Defensive Behaviors Driven by a Hypothalamic-Ventral Midbrain Circuit. eNeuro 2019; 6:ENEURO.0156-19.2019. [PMID: 31331938 PMCID: PMC6664144 DOI: 10.1523/eneuro.0156-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
The paraventricular hypothalamus (PVH) regulates stress, feeding behaviors and other homeostatic processes, but whether PVH also drives defensive states remains unknown. Here we showed that photostimulation of PVH neurons in mice elicited escape jumping, a typical defensive behavior. We mapped PVH outputs that densely terminate in the ventral midbrain (vMB) area, and found that activation of the PVH→vMB circuit produced profound defensive behavioral changes, including escape jumping, hiding, hyperlocomotion, and learned aversion. Electrophysiological recordings showed excitatory postsynaptic input onto vMB neurons via PVH fiber activation, and in vivo studies demonstrated that glutamate transmission from PVH→vMB was required for the evoked behavioral responses. Photostimulation of PVH→vMB fibers induced cFos expression mainly in non-dopaminergic neurons. Using a dual optogenetic-chemogenetic strategy, we further revealed that escape jumping and hiding were partially contributed by the activation of midbrain glutamatergic neurons. Taken together, our work unveils a hypothalamic-vMB circuit that encodes defensive properties, which may be implicated in stress-induced defensive responses.
Collapse
Affiliation(s)
- Leandra R Mangieri
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
- Graduate Program in Neuroscience of MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Zhiying Jiang
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yungang Lu
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yuanzhong Xu
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Ryan M Cassidy
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
- Graduate Program in Neuroscience of MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Nicholas Justice
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
- Graduate Program in Neuroscience of MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Benjamin R Arenkiel
- Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
- Graduate Program in Neuroscience of MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
- Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
42
|
Wang Z, Yang L, Qu S, Zhang C. CRISPR-mediated gene editing to rescue haploinsufficient obesity syndrome. Protein Cell 2019; 10:705-708. [PMID: 31124015 PMCID: PMC6776488 DOI: 10.1007/s13238-019-0635-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Zhifeng Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Research & Development Department, Sinoneural Cell and Gene Engineering Holdings Co., Ltd., Shanghai, China
| | - Liu Yang
- Department of Endocrinology and Metabolism, National Metabolic Management Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, National Metabolic Management Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
43
|
Li MM, Madara JC, Steger JS, Krashes MJ, Balthasar N, Campbell JN, Resch JM, Conley NJ, Garfield AS, Lowell BB. The Paraventricular Hypothalamus Regulates Satiety and Prevents Obesity via Two Genetically Distinct Circuits. Neuron 2019; 102:653-667.e6. [PMID: 30879785 DOI: 10.1016/j.neuron.2019.02.028] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/02/2019] [Accepted: 02/15/2019] [Indexed: 12/15/2022]
Abstract
SIM1-expressing paraventricular hypothalamus (PVH) neurons are key regulators of energy balance. Within the PVHSIM1 population, melanocortin-4 receptor-expressing (PVHMC4R) neurons are known to regulate satiety and bodyweight, yet they account for only half of PVHSIM1 neuron-mediated regulation. Here we report that PVH prodynorphin-expressing (PVHPDYN) neurons, which notably lack MC4Rs, function independently and additively with PVHMC4R neurons to account for the totality of PVHSIM1 neuron-mediated satiety. Moreover, PVHPDYN neurons are necessary for prevention of obesity in an independent but equipotent manner to PVHMC4R neurons. While PVHPDYN and PVHMC4R neurons both project to the parabrachial complex (PB), they synaptically engage distinct efferent nodes, the pre-locus coeruleus (pLC), and central lateral parabrachial nucleus (cLPBN), respectively. PB-projecting PVHPDYN neurons, like PVHMC4R neurons, receive input from interoceptive ARCAgRP neurons, respond to caloric state, and are sufficient and necessary to control food intake. This expands the CNS satiety circuitry to include two non-overlapping PVH to hindbrain circuits.
Collapse
Affiliation(s)
- Monica M Li
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jennifer S Steger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael J Krashes
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nina Balthasar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - John N Campbell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jon M Resch
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Nicholas J Conley
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alastair S Garfield
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK.
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Nyamugenda E, Trentzsch M, Russell S, Miles T, Boysen G, Phelan KD, Baldini G. Injury to hypothalamic Sim1 neurons is a common feature of obesity by exposure to high-fat diet in male and female mice. J Neurochem 2019; 149:73-97. [PMID: 30615192 DOI: 10.1111/jnc.14662] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/16/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022]
Abstract
The hypothalamus is essential for regulation of energy homeostasis and metabolism. Feeding hypercaloric, high-fat (HF) diet induces hypothalamic arcuate nucleus injury and alters metabolism more severely in male than in female mice. The site(s) and extent of hypothalamic injury in male and female mice are not completely understood. In the paraventricular nucleus (PVN) of the hypothalamus, single-minded family basic helix-loop helix transcription factor 1 (Sim1) neurons are essential to control energy homeostasis. We tested the hypothesis that exposure to HF diet induces injury to Sim1 neurons in the PVN of male and female mice. Mice expressing membrane-bound enhanced green fluorescent protein (mEGFP) in Sim1 neurons (Sim1-Cre:Rosa-mEGFP mice) were generated to visualize the effects of exposure to HF diet on these neurons. Male and female Sim1-Cre:Rosa-mEGFP mice exposed to HF diet had increased weight, hyperleptinemia, and developed hepatosteatosis. In male and female mice exposed to HF diet, expression of mEGFP was reduced by > 40% in Sim1 neurons of the PVN, an effect paralleled by cell apoptosis and neuronal loss, but not by microgliosis. In the arcuate nucleus of the Sim1-Cre:Rosa-mEGFP male mice, there was decreased alpha-melanocyte-stimulating hormone in proopiomelanocortin neurons projecting to the PVN, with increased cell apoptosis, neuronal loss, and microgliosis. These defects were undetectable in the arcuate nucleus of female mice exposed to the HF diet. Thus, injury to Sim1 neurons of the PVN is a shared feature of exposure to HF diet in mice of both sexes, while injury to proopiomelanocortin neurons in arcuate nucleus is specific to male mice. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Eugene Nyamugenda
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Marcus Trentzsch
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Susan Russell
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Tiffany Miles
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gunnar Boysen
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,The Winthrop P Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kevin D Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
45
|
Qin C, Li J, Tang K. The Paraventricular Nucleus of the Hypothalamus: Development, Function, and Human Diseases. Endocrinology 2018; 159:3458-3472. [PMID: 30052854 DOI: 10.1210/en.2018-00453] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/16/2018] [Indexed: 02/08/2023]
Abstract
The paraventricular nucleus of the hypothalamus (PVH), located in the ventral diencephalon adjacent to the third ventricle, is a highly conserved brain region present in species from zebrafish to humans. The PVH is composed of three main types of neurons, magnocellular, parvocellular, and long-projecting neurons, which play imperative roles in the regulation of energy balance and various endocrinological activities. In this review, we focus mainly on recent findings about the early development of the hypothalamus and the PVH, the functions of the PVH in the modulation of energy homeostasis and in the hypothalamus-pituitary system, and human diseases associated with the PVH, such as obesity, short stature, hypertension, and diabetes insipidus. Thus, the investigations of the PVH will benefit not only understanding of the development of the central nervous system but also the etiology of and therapy for human diseases.
Collapse
Affiliation(s)
- Cheng Qin
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi, China
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Jiaheng Li
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi, China
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Ke Tang
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Xia JY, Sun K, Hepler C, Ghaben AL, Gupta RK, An YA, Holland WL, Morley TS, Adams AC, Gordillo R, Kusminski CM, Scherer PE. Acute loss of adipose tissue-derived adiponectin triggers immediate metabolic deterioration in mice. Diabetologia 2018; 61:932-941. [PMID: 29224189 PMCID: PMC5844860 DOI: 10.1007/s00125-017-4516-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/03/2017] [Indexed: 01/08/2023]
Abstract
AIM/HYPOTHESIS Adiponectin (APN), a circulating hormone secreted by mature adipocytes, has been extensively studied because it has beneficial metabolic effects. While many studies have focused on the congenital loss of APN and its effects on systemic body glucose and lipid metabolism, little is known about the effects triggered by acute loss of APN in the adult mouse. We anticipated that genetically induced acute depletion of APN in adult mice would have a more profound effect on systemic metabolic health than congenital deletion of Adipoq, the gene encoding APN, with its associated potential for adaptive responses that may mask the phenotypes. METHODS Mice carrying loxP-flanked regions of Adipoq were generated and bred to the Adipoq (APN) promoter-driven reverse tetracycline-controlled transactivator (rtTA) (APN-rtTA) gene and a tet-responsive Cre line (TRE-Cre) to achieve acute depletion of APN. Upon acute removal of APN in adult mice, systemic glucose and lipid homeostasis were assessed under basal and insulinopenic conditions. RESULTS The acute depletion of APN results in more severe systemic insulin resistance and hyperlipidaemia than in mice with congenital loss of APN. Furthermore, the acute depletion of APN in adult mice results in a much more dramatic reduction in survival rate, with 50% of inducible knockouts dying in the first 5 days under insulinopenic conditions compared with 0% of congenital Adipoq knockout mice under similar conditions. CONCLUSIONS/INTERPRETATION Acute systemic removal of APN results in a much more negative metabolic phenotype compared with congenital knockout of Adipoq. Specifically, our data demonstrate that acute depletion of APN is especially detrimental to lipid homeostasis, both under basal and insulinopenic conditions. This suggests that compensatory mechanisms exist in congenital knockout mice that offset some of the metabolic actions covered by APN.
Collapse
Affiliation(s)
- Jonathan Y Xia
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
- McGaw Medical Center of Northwestern University, Department of Internal Medicine, Chicago, IL, USA
| | - Kai Sun
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chelsea Hepler
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Alexandra L Ghaben
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Thomas S Morley
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Andrew C Adams
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA.
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
47
|
Understanding melanocortin-4 receptor control of neuronal circuits: Toward novel therapeutics for obesity syndrome. Pharmacol Res 2018; 129:10-19. [DOI: 10.1016/j.phrs.2018.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 01/25/2023]
|
48
|
A neural basis for antagonistic control of feeding and compulsive behaviors. Nat Commun 2018; 9:52. [PMID: 29302029 PMCID: PMC5754347 DOI: 10.1038/s41467-017-02534-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023] Open
Abstract
Abnormal feeding often co-exists with compulsive behaviors, but the underlying neural basis remains unknown. Excessive self-grooming in rodents is associated with compulsivity. Here, we show that optogenetically manipulating the activity of lateral hypothalamus (LH) projections targeting the paraventricular hypothalamus (PVH) differentially promotes either feeding or repetitive self-grooming. Whereas selective activation of GABAergic LH→PVH inputs induces feeding, activation of glutamatergic inputs promotes self-grooming. Strikingly, targeted stimulation of GABAergic LH→PVH leads to rapid and reversible transitions to feeding from induced intense self-grooming, while activating glutamatergic LH→PVH or PVH neurons causes rapid and reversible transitions to self-grooming from voracious feeding induced by fasting. Further, specific inhibition of either LH→PVH GABAergic action or PVH neurons reduces self-grooming induced by stress. Thus, we have uncovered a parallel LH→PVH projection circuit for antagonistic control of feeding and self-grooming through dynamic modulation of PVH neuron activity, revealing a common neural pathway that underlies feeding and compulsive behaviors.
Collapse
|
49
|
Xu J, Bartolome CL, Kong D. Synaptic Regulation of Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:49-77. [PMID: 30390285 DOI: 10.1007/978-981-13-1286-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neurons in the brain, particularly those in the hypothalamus, are essential for the maintenance of whole-body metabolic homeostasis. Dysfunctions or dysregulations of them can result in various metabolic diseases, including eating disorders, obesity, and diabetes. In addition to hormonal and peptidergic regulation, accumulating evidence has shown that these neurons are subject to synaptic regulation, which has been largely overlooked. In this chapter, we focus on synaptic neurotransmission of hypothalamic neurons and summarize current knowledge of synaptic plasticity in the maintenance of energy balance. Synaptic modulation engaged by circulating hormones is also discussed.
Collapse
Affiliation(s)
- Jie Xu
- Department of Neuroscience, Tufts University School of Medicine, Tufts University, Boston, MA, USA.,Program of Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Christopher L Bartolome
- Department of Neuroscience, Tufts University School of Medicine, Tufts University, Boston, MA, USA.,Program of Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Dong Kong
- Department of Neuroscience, Tufts University School of Medicine, Tufts University, Boston, MA, USA. .,Program of Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
50
|
Semple E, Hill JW. Sim1 Neurons Are Sufficient for MC4R-Mediated Sexual Function in Male Mice. Endocrinology 2018; 159:439-449. [PMID: 29059347 PMCID: PMC5761591 DOI: 10.1210/en.2017-00488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/12/2017] [Indexed: 12/24/2022]
Abstract
Sexual dysfunction is a poorly understood condition that affects up to one-third of men around the world. Existing treatments that target the periphery do not work for all men. Previous studies have shown that central melanocortins, which are released by pro-opiomelanocortin neurons in the arcuate nucleus of the hypothalamus, can lead to male erection and increased libido. Several studies specifically implicate the melanocortin 4 receptor (MC4R) in the central control of sexual function, but the specific neural circuitry involved is unknown. We hypothesized that single-minded homolog 1 (Sim1) neurons play an important role in the melanocortin-mediated regulation of male sexual behavior. To test this hypothesis, we examined the sexual behavior of mice expressing MC4R only on Sim1-positive neurons (tbMC4Rsim1 mice) in comparison with tbMC4R null mice and wild-type controls. In tbMC4Rsim1 mice, MC4R reexpression was found in the medial amygdala and paraventricular nucleus of the hypothalamus. These mice were paired with sexually experienced females, and their sexual function and behavior was scored based on mounting, intromission, and ejaculation. tbMC4R null mice showed a longer latency to mount, a reduced intromission efficiency, and an inability to reach ejaculation. Expression of MC4R only on Sim1 neurons reversed the sexual deficits seen in tbMC4R null mice. This study implicates melanocortin signaling via the MC4R on Sim1 neurons in the central control of male sexual behavior.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/metabolism
- Amygdala/pathology
- Animals
- Arcuate Nucleus of Hypothalamus/drug effects
- Arcuate Nucleus of Hypothalamus/metabolism
- Arcuate Nucleus of Hypothalamus/pathology
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Copulation/drug effects
- Crosses, Genetic
- Fertility Agents, Male/administration & dosage
- Fertility Agents, Male/therapeutic use
- Heterozygote
- Infertility, Male/drug therapy
- Infertility, Male/metabolism
- Infertility, Male/pathology
- Injections, Intraventricular
- Male
- Mice, Knockout
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
- Organ Specificity
- Paraventricular Hypothalamic Nucleus/drug effects
- Paraventricular Hypothalamic Nucleus/metabolism
- Paraventricular Hypothalamic Nucleus/pathology
- Random Allocation
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Repressor Proteins/metabolism
- Sexual Behavior, Animal/drug effects
- alpha-MSH/administration & dosage
- alpha-MSH/therapeutic use
Collapse
Affiliation(s)
- Erin Semple
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43606
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43606
| |
Collapse
|