1
|
Chen YF, Tsao CY, Chen YT, Chang HC, Li WY, Chiang JL, Chen CFF, Chen CH, Gau SSF, Lee KY, Lee LJ, Wang YC. Altered odor perception in Dlgap2 mutant mice, a mouse model of autism spectrum disorder. Behav Brain Res 2025; 480:115365. [PMID: 39631506 DOI: 10.1016/j.bbr.2024.115365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Olfactory dysfunction has been observed in patients with Autism Spectrum Disorder (ASD). A microdeletion at the 8p23 terminal regions of chromosome 8p23 was identified in a Taiwanese patient with ASD, suggesting a potential association with mutations in the DLGAP2 gene. DLGAP2 is expressed in the olfactory bulb in rodents. The current study investigated olfactory phenotypes of Dlgap2 mutant mice. The results indicated that odor detection capabilities were comparable between wild-type (WT) and Dlgap2 mutant mice. However, homozygous mutant (Homo) mice showed less interest in sniffing odors of banana and almond but greater sniffing activity in response to bedding from unfamiliar cages. Notably, exposure to banana odor elicited significant c-fos expression in most olfaction-related brain regions of WT mice, while Homo mice did not show much increase in c-fos levels in major olfactory areas, which may correlate with their diminished sniffing behavior. Bedding stimuli induced pronounced c-fos expression in WT brains and some olfaction-related regions, including the olfactory bulb, amygdala, hypothalamus, and medial prefrontal cortex, in Homo mice. These mutants may still process olfactory signals from the bedding through a relatively narrow channel, which might elicit their interest, leading to increased sniffing behaviors that may compensate for their olfactory deficits. The DLGAP2 protein was absent in the olfactory bulb of Homo mice, and the levels of PSD95 and CaMKIIβ were also affected, indicating alterations in synaptic transmission and signaling within the olfactory system. This study evaluated olfactory perception in a mouse model of ASD, which may advance diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yu-Fu Chen
- Department of Neurology, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yuh-Tarng Chen
- Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ho-Ching Chang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wai-Yu Li
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jui-Lin Chiang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chien-Fu Fred Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Susan Shur-Fen Gau
- Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Kuang-Yung Lee
- Department of Neurology, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan; Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Yu-Chun Wang
- Department of Otolaryngology, Head and Neck Surgery, Chi-Mei Medical Center, Tainan, Taiwan.
| |
Collapse
|
2
|
Zizzari P, Castellanos-Jankiewicz A, Yagoub S, Simon V, Clark S, Maître M, Dupuy N, Leste-Lasserre T, Gonzales D, Schoonjans K, Fénelon VS, Cota D. TGR5 receptors in SF1-expressing neurons of the ventromedial hypothalamus regulate glucose homeostasis. Mol Metab 2025; 91:102071. [PMID: 39603503 PMCID: PMC11650306 DOI: 10.1016/j.molmet.2024.102071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE Steroidogenic factor-1 (SF1) neurons of the ventromedial hypothalamus play key roles in the regulation of food intake, body weight and glucose metabolism. The bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) is expressed in the hypothalamus, where it determines some of the actions of bile acids on food intake and body weight through still poorly defined neuronal mechanisms. Here, we examined the role of TGR5 in SF1 neurons in the regulation of energy balance and glucose metabolism. METHODS We used a genetic approach combined with metabolic phenotyping and molecular analyses to establish the effect of TGR5 deletion in SF1 neurons on meal pattern, body weight, body composition, energy expenditure and use of energy substrates as well as on possible changes in glucose handling and insulin sensitivity. RESULTS Our findings reveal that TGR5 in SF1 neurons does not play a major role in the regulation of food intake or body weight under standard chow, but it is involved in the adaptive feeding response to the acute exposure to cold or to a hypercaloric, high-fat diet, without changes in energy expenditure. Notably, TGR5 in SF1 neurons hinder glucose metabolism, since deletion of the receptor improves whole-body glucose uptake through heightened insulin signaling in the hypothalamus and in the brown adipose tissue. CONCLUSIONS TGR5 in SF1 neurons favours satiety by differently modifying the meal pattern in response to specific metabolic cues. These studies also reveal a novel key function for TGR5 in SF1 neurons in the regulation of whole-body insulin sensitivity, providing new insight into the role played by neuronal TGR5 in the regulation of metabolism.
Collapse
Affiliation(s)
- Philippe Zizzari
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | | | - Selma Yagoub
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Vincent Simon
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Samantha Clark
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Marlene Maître
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Nathalie Dupuy
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | | | - Delphine Gonzales
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Kristina Schoonjans
- Institute of Bioengineering, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Valérie S Fénelon
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
3
|
Nasanbuyan N, Yoshida M, Inutsuka A, Takayanagi Y, Kato S, Hidema S, Nishimori K, Kobayashi K, Onaka T. Differential Functions of Oxytocin Receptor-Expressing Neurons in the Ventromedial Hypothalamus in Social Stress Responses: Induction of Adaptive and Maladaptive Coping Behaviors. Biol Psychiatry 2024:S0006-3223(24)01615-9. [PMID: 39343339 DOI: 10.1016/j.biopsych.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The flexibility to adjust actions and attitudes in response to varying social situations is a fundamental aspect of adaptive social behavior. Adaptive social behaviors influence an individual's vulnerability to social stress. While it has been proposed that oxytocin is a facilitator of active coping behaviors during social stress, the exact mechanisms remain unknown. METHODS Using a social defeat stress paradigm in male mice, we identified the distribution of oxytocin receptor (OXTR)-expressing neurons in the ventrolateral part of the ventromedial hypothalamus (vlVMH) that are activated during stress by detection of c-Fos protein expression. We then investigated the role of vlVMH OXTR-expressing neurons in social defeat stress responses by chemogenetic methods or deletion of local OXTRs. The social defeat posture was measured for quantification of adaptive social behavior during repeated social stress. RESULTS Social defeat stress activated OXTR-expressing neurons rather than estrogen receptor 1-expressing neurons in the rostral vlVMH. OXTR-expressing neurons in the vlVMH were glutamatergic. Chemogenetic activation of vlVMH OXTR-expressing neurons facilitated exhibition of the social defeat posture during exposure to social stress, while local OXTR deletion suppressed it. In contrast, overactivation of vlVMH-OXTR neurons induced generalized social avoidance after exposure to chronic social defeat stress. Neural circuits for the social defeat posture centered on OXTR-expressing neurons were identified by viral tracers and c-Fos mapping. CONCLUSIONS vlVMH OXTR-expressing neurons are a functionally unique population of neurons that promote active coping behavior during social stress, but their excessive and repetitive activation under chronic social stress impairs subsequent social behavior.
Collapse
Affiliation(s)
- Naranbat Nasanbuyan
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan
| | - Masahide Yoshida
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan.
| | - Ayumu Inutsuka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shizu Hidema
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuhiko Nishimori
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
4
|
Otten J, Dan S, Rostin L, Profetto AE, Lardenoije R, Klengel T. Spatial transcriptomics reveals modulation of transcriptional networks across brain regions after auditory threat conditioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614979. [PMID: 39386587 PMCID: PMC11463379 DOI: 10.1101/2024.09.25.614979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Prior research has demonstrated genome-wide transcriptional changes related to fear and anxiety across species, often focusing on individual brain regions or cell types. However, the extent of gene expression differences across brain regions and how these changes interact at the level of transcriptional connectivity remains unclear. To address this, we performed spatial transcriptomics RNAseq analyses in an auditory threat conditioning paradigm in mice. We generated a spatial transcriptomic atlas of a coronal mouse brain section covering cortical and subcortical regions, corresponding to histologically defined regions. Our finding revealed widespread transcriptional responses across all brain regions examined, particularly in the medial and lateral habenula, and the choroid plexus. Network analyses highlighted altered transcriptional connectivity between cortical and subcortical regions, emphasizing the role of steroidogenic factor 1. These results provide new insights into the transcriptional networks involved in auditory threat conditioning, enhancing our understanding of molecular and neural mechanisms underlying fear and anxiety disorders.
Collapse
|
5
|
Li Y, Deng Y, Zhang Y, Xu D, Zhang X, Li Y, Li Y, Chen M, Wang Y, Zhang J, Wang L, Cang Y, Cao P, Bi L, Xu H. Distinct glutamatergic projections of the posteroventral medial amygdala play different roles in arousal and anxiety. JCI Insight 2024; 9:e176329. [PMID: 38842948 PMCID: PMC11383360 DOI: 10.1172/jci.insight.176329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 06/05/2024] [Indexed: 08/13/2024] Open
Abstract
Sleep disturbance usually accompanies anxiety disorders and exacerbates their incidence rates. The precise circuit mechanisms remain poorly understood. Here, we found that glutamatergic neurons in the posteroventral medial amygdala (MePVGlu neurons) are involved in arousal and anxiety-like behaviors. Excitation of MePVGlu neurons not only promoted wakefulness but also increased anxiety-like behaviors. Different projections of MePVGlu neurons played various roles in regulating anxiety-like behaviors and sleep-wakefulness. MePVGlu neurons promoted wakefulness through the MePVGlu/posteromedial cortical amygdaloid area (PMCo) pathway and the MePVGlu/bed nucleus of the stria terminals (BNST) pathway. In contrast, MePVGlu neurons increased anxiety-like behaviors through the MePVGlu/ventromedial hypothalamus (VMH) pathway. Chronic sleep disturbance increased anxiety levels and reduced reparative sleep, accompanied by the enhanced excitability of MePVGlu/PMCo and MePVGlu/VMH circuits but suppressed responses of glutamatergic neurons in the BNST. Inhibition of the MePVGlu neurons could rescue chronic sleep deprivation-induced phenotypes. Our findings provide important circuit mechanisms for chronic sleep disturbance-induced hyperarousal response and obsessive anxiety-like behavior and are expected to provide a promising strategy for treating sleep-related psychiatric disorders and insomnia.
Collapse
Affiliation(s)
- Ying Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuchen Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yifei Zhang
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Center for Pathology and Molecular Diagnostics
| | - Dan Xu
- Department of Nuclear Medicine, and
| | - Xuefen Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yue Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yidan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ming Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuxin Wang
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Center for Pathology and Molecular Diagnostics
| | - Jiyan Zhang
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Center for Pathology and Molecular Diagnostics
| | - Like Wang
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Center for Pathology and Molecular Diagnostics
| | - Yufeng Cang
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Center for Pathology and Molecular Diagnostics
| | - Peng Cao
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Linlin Bi
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Center for Pathology and Molecular Diagnostics
- Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Shao J, Chen Y, Gao D, Liu Y, Hu N, Yin L, Zhang X, Yang F. Ventromedial hypothalamus relays chronic stress inputs and exerts bidirectional regulation on anxiety state and related sympathetic activity. Front Cell Neurosci 2023; 17:1281919. [PMID: 38161999 PMCID: PMC10755867 DOI: 10.3389/fncel.2023.1281919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Chronic stress can induce negative emotion states, including anxiety and depression, leading to sympathetic overactivation and disturbed physiological homeostasis in peripheral tissues. While anxiety-related neural circuitry integrates chronic stress information and modulates sympathetic nervous system (SNS) activity, the critical nodes linking anxiety and sympathetic activity still need to be clarified. In our previous study, we demonstrated that the ventromedial hypothalamus (VMH) is involved in integrating chronic stress inputs and exerting influence on sympathetic activity. However, the underlying synaptic and electrophysiological mechanisms remain elusive. In this study, we combined in vitro electrophysiological recordings, behavioral tests, optogenetic manipulations, and SNS activity analyses to explore the role of VMH in linking anxiety emotion and peripheral SNS activity. Results showed that the VMH played an important role in bidirectionally regulating anxiety-like behavior and peripheral sympathetic excitation. Chronic stress enhanced excitatory inputs into VMH neurons by strengthening the connection with the paraventricular hypothalamus (PVN), hence promoting anxiety and sympathetic tone outflow, an important factor contributing to the development of metabolic imbalance in peripheral tissues and cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Shao
- Department of Nephrology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yan Chen
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Dashuang Gao
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Yunhui Liu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Nan Hu
- Department of Nephrology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Lianghong Yin
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xinzhou Zhang
- Department of Nephrology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Fan Yang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| |
Collapse
|
7
|
Li Y, Li Y, Zhang X, Li Y, Liu Y, Xu H. CaMKIIa Neurons of the Ventromedial Hypothalamus Mediate Wakefulness and Anxiety-like Behavior. Neurochem Res 2023:10.1007/s11064-023-03925-9. [PMID: 37014492 DOI: 10.1007/s11064-023-03925-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 04/05/2023]
Abstract
Insomnia and anxiety are two common and closely related clinical problems that pose a threat to individuals' physical and mental well-being. There is a possibility that some nuclei and neural circuits in the brain are shared by both insomnia and anxiety. In the present study, using a combination of chemogenetics, optogenetics, polysomnographic recordings and the classic tests of anxiety-like behaviors, we verified that the calmodulin-dependent protein kinase II alpha (CaMKIIa) neurons of the ventromedial hypothalamus (VMH) are involved in the regulation of both wakefulness and anxiety. Chemogenetic manipulation of the VMH CaMKIIa neurons elicited an apparent increase in wakefulness during activation, whereas inhibition decreased wakefulness mildly. It substantiated that the VMH CaMKIIa neurons contribute to wakefulness. Then in millisecond-scale control of neuronal activity, short-term and long-term optogenetic activation induced the initiation and maintenance of wakefulness, respectively. We also observed that mice reduced exploratory behaviors in classic anxiety tests while activating the VMH CaMKIIa neurons and were anxiolytic while inhibiting. Additionally, photostimulation of the VMH CaMKIIa axons in the paraventricular hypothalamus (PVH) mediated wakefulness and triggered anxiety-like behaviors as well. In conclusion, our results demonstrate that the VMH participates in the control of wakefulness and anxiety, and offer a neurological explanation for insomnia and anxiety, which may be valuable for therapeutic interventions such as medication and transcranial magnetic stimulation.
Collapse
Affiliation(s)
- Yidan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, 430071, China
| | - Yue Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, 430071, China
| | - Xuefen Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, 430071, China
| | - Ying Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, 430071, China
| | - Yanchao Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, 430071, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, 430071, China.
| |
Collapse
|
8
|
Zhang XH, Shen CL, Wang XY, Xiong WF, Shang X, Tang LY, Zhang HX, Wan YH, Wu YB, Fei J, Yi QZ, Wang ZG. Increased Anxiety-like Behaviors in Adhesion G protein-coupled receptor A1 -/- Male But Not Female Mice are Attributable to Elevated Neuron Dendritic Density, Upregulated Postsynaptic Density Protein 95 Expression, and Abnormal Activation of the Phosphatidylinositol 3 Kinase/Protein Kinase B/Glycogen Synthase Kinase-3 and Methyl Ethyl Ketone/Extracellular Signal Regulated Kinase Pathways. Neuroscience 2022; 503:131-145. [PMID: 36115515 DOI: 10.1016/j.neuroscience.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022]
Abstract
Adhesion G protein-coupled receptor A1 (ADGRA1) belongs to the G protein-coupled receptor (GPCR) family, and its physiological function remains largely unknown. We found that Adgra1 is highly and exclusively expressed in the brain, suggesting that Adgra1 may be involved in the regulation of neurological behaviors including anxiety, depression, learning and memory. To this end, we comprehensively analyzed the potential role of ADGRA1 in the neurobehaviors of mice by comparing Adgra1-/- and their wild-type (wt) littermates. We found that Adgra1-/- male but not female mice exhibited elevated anxiety levels in the open field, elevated plus maze, and light-dark box tests, with normal depression levels in the tail-suspension and forced-swim tests, and comparable learning and memory abilities in the Morris water maze, Y maze, fear condition, and step-down avoidance tests. Further studies showed that ADGRA1 deficiency resulted in higher dendritic branching complexity and spine density as evidenced by elevated expression levels of SYN and PSD95 in amygdalae-of male mice. Finally, we found that PI3K/AKT/GSK-3β and MEK/ERK in amygdalae of Adgra1-deficient male mice were aberrantly activated when compared to wt male mice. Together, our findings reveal an important suppressive role of ADGRA1 in anxiety control and synaptic function by regulating the PI3K/AKT/GSK-3β and MEK/ERK pathways in amygdalae of male mice, implicating a potential, therapeutic application in novel anti-anxiety drug development.
Collapse
Affiliation(s)
- Xiao-Hong Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Chun-Ling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Xi-Yi Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China; Department of Obstetrics and Gynecology, Tang-Du Hospital Affiliated to the Fourth Military Medical University, Xi'an 710038, China.
| | - Wen-Feng Xiong
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Xuan Shang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Ling-Yun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Hong-Xin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Ying-Han Wan
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| | - You-Bing Wu
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| | - Jian Fei
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| | - Qi-Zhong Yi
- Psychological Medical Center, The First Hospital affiliated to Xin Jiang Medical University, Urumqi 830054, China; Xin Jiang Clinical Research Center for Mental Health, Urumqi 830054, China.
| | - Zhu-Gang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China; Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| |
Collapse
|
9
|
Wang Q, Zhang B, Stutz B, Liu ZW, Horvath TL, Yang X. Ventromedial hypothalamic OGT drives adipose tissue lipolysis and curbs obesity. SCIENCE ADVANCES 2022; 8:eabn8092. [PMID: 36044565 PMCID: PMC9432828 DOI: 10.1126/sciadv.abn8092] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/14/2022] [Indexed: 05/31/2023]
Abstract
The ventromedial hypothalamus (VMH) is known to regulate body weight and counterregulatory response. However, how VMH neurons regulate lipid metabolism and energy balance remains unknown. O-linked β-d-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation), catalyzed by O-GlcNAc transferase (OGT), is considered a cellular sensor of nutrients and hormones. Here, we report that genetic ablation of OGT in VMH neurons inhibits neuronal excitability. Mice with VMH neuron-specific OGT deletion show rapid weight gain, increased adiposity, and reduced energy expenditure, without significant changes in food intake or physical activity. The obesity phenotype is associated with adipocyte hypertrophy and reduced lipolysis of white adipose tissues. In addition, OGT deletion in VMH neurons down-regulates the sympathetic activity and impairs the sympathetic innervation of white adipose tissues. These findings identify OGT in the VMH as a homeostatic set point that controls body weight and underscore the importance of the VMH in regulating lipid metabolism through white adipose tissue-specific innervation.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Bichen Zhang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Bernardo Stutz
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L. Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiaoyong Yang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
10
|
Rawlinson S, Reichenbach A, Clarke RE, Nuñez-Iglesias J, Dempsey H, Lockie SH, Andrews ZB. In Vivo Photometry Reveals Insulin and 2-Deoxyglucose Maintain Prolonged Inhibition of VMH Vglut2 Neurons in Male Mice. Endocrinology 2022; 163:6631280. [PMID: 35788848 DOI: 10.1210/endocr/bqac095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Indexed: 11/19/2022]
Abstract
The ventromedial hypothalamic (VMH) nucleus is a well-established hub for energy and glucose homeostasis. In particular, VMH neurons are thought to be important for initiating the counterregulatory response to hypoglycemia, and ex vivo electrophysiology and immunohistochemistry data indicate a clear role for VMH neurons in sensing glucose concentration. However, the temporal response of VMH neurons to physiologically relevant changes in glucose availability in vivo has been hampered by a lack of available tools for measuring neuronal activity over time. Since the majority of neurons within the VMH are glutamatergic and can be targeted using the vesicular glutamate transporter Vglut2, we expressed cre-dependent GCaMP7s in Vglut2 cre mice and examined the response profile of VMH to intraperitoneal injections of glucose, insulin, and 2-deoxyglucose (2DG). We show that reduced available glucose via insulin-induced hypoglycemia and 2DG-induced glucoprivation, but not hyperglycemia induced by glucose injection, inhibits VMH Vglut2 neuronal population activity in vivo. Surprisingly, this inhibition was maintained for at least 45 minutes despite prolonged hypoglycemia and initiation of a counterregulatory response. Thus, although VMH stimulation, via pharmacological, electrical, or optogenetic approaches, is sufficient to drive a counterregulatory response, our data suggest VMH Vglut2 neurons are not the main drivers required to do so, since VMH Vglut2 neuronal population activity remains suppressed during hypoglycemia and glucoprivation.
Collapse
Affiliation(s)
- Sasha Rawlinson
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Alex Reichenbach
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Rachel E Clarke
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Juan Nuñez-Iglesias
- Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Harry Dempsey
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Sarah H Lockie
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
11
|
Velasco ER, Florido A, Flores Á, Senabre E, Gomez-Gomez A, Torres A, Roca A, Norrholm S, Newman EL, Das P, Ross RA, Lori A, Pozo OJ, Ressler KJ, Garcia-Esteve LL, Jovanovic T, Andero R. PACAP-PAC1R modulates fear extinction via the ventromedial hypothalamus. Nat Commun 2022; 13:4374. [PMID: 35902577 PMCID: PMC9334354 DOI: 10.1038/s41467-022-31442-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/16/2022] [Indexed: 12/14/2022] Open
Abstract
Exposure to traumatic stress can lead to fear dysregulation, which has been associated with posttraumatic stress disorder (PTSD). Previous work showed that a polymorphism in the PACAP-PAC1R (pituitary adenylate cyclase-activating polypeptide) system is associated with PTSD risk in women, and PACAP (ADCYAP1)-PAC1R (ADCYAP1R1) are highly expressed in the hypothalamus. Here, we show that female mice subjected to acute stress immobilization (IMO) have fear extinction impairments related to Adcyap1 and Adcyap1r1 mRNA upregulation in the hypothalamus, PACAP-c-Fos downregulation in the Medial Amygdala (MeA), and PACAP-FosB/ΔFosB upregulation in the Ventromedial Hypothalamus dorsomedial part (VMHdm). DREADD-mediated inhibition of MeA neurons projecting to the VMHdm during IMO rescues both PACAP upregulation in VMHdm and the fear extinction impairment. We also found that women with the risk genotype of ADCYAP1R1 rs2267735 polymorphism have impaired fear extinction.
Collapse
Affiliation(s)
- E R Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - A Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Á Flores
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - E Senabre
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
| | - A Gomez-Gomez
- Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - A Torres
- Perinatal Mental health Unit, Department of Psychiatry and Clinical Psychology, Institute of Neuroscience, Hospital Clínic, IDIBAPS, Barcelona, Spain
- Programme for the Prevention and Treatment of Psychic Effects in Sexually Assaulted Women. Hospital Clínic de Barcelona, Barcelona, Spain
| | - A Roca
- Perinatal Mental health Unit, Department of Psychiatry and Clinical Psychology, Institute of Neuroscience, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - S Norrholm
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University, Detroit, MI, USA
| | - E L Newman
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - P Das
- Department of Neuroscience, Albert Einstein College of Medicine, Psychiatry Research Institute of Montefiore and Einstein, New York, NY, USA
| | - R A Ross
- Department of Neuroscience, Albert Einstein College of Medicine, Psychiatry Research Institute of Montefiore and Einstein, New York, NY, USA
| | - A Lori
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
- American Cancer Society, Inc., Atlanta, GA, USA
| | - O J Pozo
- Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - K J Ressler
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - L L Garcia-Esteve
- Perinatal Mental health Unit, Department of Psychiatry and Clinical Psychology, Institute of Neuroscience, Hospital Clínic, IDIBAPS, Barcelona, Spain
- Programme for the Prevention and Treatment of Psychic Effects in Sexually Assaulted Women. Hospital Clínic de Barcelona, Barcelona, Spain
| | - T Jovanovic
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University, Detroit, MI, USA
| | - R Andero
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
12
|
Sherman SB, Harberson M, Rashleigh R, Gupta N, Powers R, Talla R, Thusu A, Hill JW. Spexin modulates molecular thermogenic profile of adipose tissue and thermoregulatory behaviors in female C57BL/6 mice. Horm Behav 2022; 143:105195. [PMID: 35580373 PMCID: PMC10150790 DOI: 10.1016/j.yhbeh.2022.105195] [Citation(s) in RCA: 3] [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: 11/11/2021] [Revised: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
Thermoregulation is the physiological process by which an animal regulates body temperature in response to its environment. It is known that galanin, a neuropeptide widely distributed throughout the central nervous system and secreted by the gut, plays a role in thermoregulatory behaviors and metabolism. We tested the ability of the novel neuropeptide spexin, which shares sequence homology to galanin, to regulate these functions in female mice. Supraphysiological levels of spexin in C57BL/6 mice did not lead to weight loss after 50 days of treatment. Behavioral analysis of long-term spexin treatment showed it decreased anxiety and increased thermoregulatory nest building, which was not observed when mice were housed at thermoneutral temperatures. Treatment also disrupted the thermogenic profile of brown and white adipose tissue, decreasing mRNA expression of Ucp1 in BAT and immunodetection of β3-adrenergic receptors in gWAT. Our results reveal novel functions for spexin as a modulator of thermoregulatory behaviors and adipose tissue metabolism.
Collapse
Affiliation(s)
- Shermel B Sherman
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, Toledo, OH 43614, United States
| | - Mitchell Harberson
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, Toledo, OH 43614, United States
| | - Rebecca Rashleigh
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, Toledo, OH 43614, United States
| | - Niraj Gupta
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Department of Bioengineering, University of Toledo, Toledo, OH 43604, United States
| | - Riley Powers
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, United States
| | - Ramya Talla
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, Toledo, OH 43614, United States
| | - Ashima Thusu
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Department of Bioengineering, University of Toledo, Toledo, OH 43604, United States
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, Toledo, OH 43614, United States.
| |
Collapse
|
13
|
Abstract
The role of central estrogen in cognitive, metabolic, and reproductive health has long fascinated the lay public and scientists alike. In the last two decades, insight into estrogen signaling in the brain and its impact on female physiology is beginning to catch up with the vast information already established for its actions on peripheral tissues. Using newer methods to manipulate estrogen signaling in hormone-sensitive brain regions, neuroscientists are now identifying the molecular pathways and neuronal subtypes required for controlling sex-dependent energy allocation. However, the immense cellular complexity of these hormone-sensitive brain regions makes it clear that more research is needed to fully appreciate how estrogen modulates neural circuits to regulate physiological and behavioral end points. Such insight is essential for understanding how natural or drug-induced hormone fluctuations across lifespan affect women's health.
Collapse
Affiliation(s)
- Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| |
Collapse
|
14
|
Cav3.1-driven bursting firing in ventromedial hypothalamic neurons exerts dual control of anxiety-like behavior and energy expenditure. Mol Psychiatry 2022; 27:2901-2913. [PMID: 35318460 PMCID: PMC9156408 DOI: 10.1038/s41380-022-01513-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/22/2022]
Abstract
The central nervous system has evolved to coordinate the regulation of both the behavior response to the external environment and homeostasis of energy expenditure. Recent studies have indicated the dorsomedial ventromedial hypothalamus (dmVMH) as an important hub that regulates both innate behavior and energy homeostasis for coping stress. However, how dmVMH neurons control neuronal firing pattern to regulate chronic stress-induced anxiety and energy expenditure remains poorly understood. Here, we found enhanced neuronal activity in VMH after chronic stress, which is mainly induced by increased proportion of burst firing neurons. This enhancement of VMH burst firing is predominantly mediated by Cav3.1 expression. Optogenetically evoked burst firing of dmVMH neurons induced anxiety-like behavior, shifted the respiratory exchange ratio toward fat oxidation, and decreased food intake, while knockdown of Cav3.1 in the dmVMH had the opposite effects, suggested that Cav 3.1 as a crucial regulator. Interestingly, we found that fluoxetine (anxiolytics) could block the increase of Cav3.1 expression to inhibit the burst firing, and then rescued the anxiety-like behaviors and energy expenditure changes. Collectively, our study first revealed an important role of Cav3.1-driven bursting firing of dmVMH neurons in the control of anxiety-like behavior and energy expenditure, and provided potential therapeutic targets for treating the chronic stress-induced emotional malfunction and metabolism disorders.
Collapse
|
15
|
Zhou Y, Liu Z, Liu Z, Zhou H, Xu X, Li Z, Chen H, Wang Y, Zhou Z, Wang M, Lai Y, Zhou L, Zhou X, Jiang H. Ventromedial Hypothalamus Activation Aggravates Hypertension Myocardial Remodeling Through the Sympathetic Nervous System. Front Cardiovasc Med 2021; 8:737135. [PMID: 34733893 PMCID: PMC8558385 DOI: 10.3389/fcvm.2021.737135] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The ventromedial hypothalamus (VMH) is an important nuclei in responding to emotional stress, and emotional stress is a risk factor for cardiovascular diseases. However, the role of the VMH in cardiovascular diseases remains unknown. This study aimed to investigate the effects and underlying mechanisms of VMH activation on hypertension related cardiac remodeling in two-kidney-one-clip (2K1C) hypertension (HTN) rats. Methods: Eighteen male Sprague-Dawley rats were injected with AAV-hSyn-hM3D(Gq) into the VMH at 0 weeks and then randomly divided into three groups: (1) sham group (sham 2K1C + saline i.p. injection); (2) HTN group (2K1C + saline i.p. injection); (3) HTN+VMH activation group (2K1C + clozapine-N-oxide i.p. injection). One week later, rats were subjected to a sham or 2K1C operation, and 2 weeks later rats were injected with clozapine-N-oxide or saline for 2 weeks. Results: In the HTN+VMH activation group, FosB expression was significantly increased in VMH sections compared with those of the other two groups. Compared to the HTN group, the HTN+VMH activation group showed significant: (1) increases in systolic blood pressure (SBP); (2) exacerbation of cardiac remodeling; and (3) increases in serum norepinephrine levels and sympathetic indices of heart rate variability. Additionally, myocardial RNA-sequencing analysis showed that VMH activation might regulate the HIF-1 and PPAR signal pathway and fatty acid metabolism. qPCR results confirmed that the relative mRNA expression of HIF-1α was increased and the PPARα and CPT-1 mRNA expression were decreased in the HTN+VMH activation group compared to the HTN group. Conclusions: VMH activation could increase SBP and aggravate cardiac remodeling possibly by sympathetic nerve activation and the HIF-1α/PPARα/CPT-1 signaling pathway might be the underlying mechanism.
Collapse
Affiliation(s)
- Yuyang Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huixin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zeyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuhong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Meng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanqiu Lai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiac Autonomic Nervous Research Center, Wuhan University, Wuhan, China.,Department of Cardiology Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
16
|
Astrocytes in the Ventromedial Hypothalamus Involve Chronic Stress-Induced Anxiety and Bone Loss in Mice. Neural Plast 2021; 2021:7806370. [PMID: 34306063 PMCID: PMC8282369 DOI: 10.1155/2021/7806370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/22/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic stress is one of the main risk factors of bone loss. While the neurons and neural circuits of the ventromedial hypothalamus (VMH) mediate bone loss induced by chronic stress, the detailed intrinsic mechanisms within the VMH nucleus still need to be explored. Astrocytes in brain regions play important roles in the regulation of metabolism and anxiety-like behavior through interactions with surrounding neurons. However, whether astrocytes in the VMH affect neuronal activity and therefore regulate chronic stress-induced anxiety and bone loss remain elusive. In this study, we found that VMH astrocytes were activated during chronic stress-induced anxiety and bone loss. Pharmacogenetic activation of the Gi and Gq pathways in VMH astrocytes reduced and increased the levels of anxiety and bone loss, respectively. Furthermore, activation of VMH astrocytes by optogenetics induced depolarization in neighboring steroidogenic factor-1 (SF-1) neurons, which was diminished by administration of N-methyl-D-aspartic acid (NMDA) receptor blocker but not by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker. These results suggest that there may be a functional "glial-neuron microcircuit" in VMH nuclei that mediates anxiety and bone loss induced by chronic stress. This study not only advances our understanding of glial cell function but also provides a potential intervention target for chronic stress-induced anxiety and bone loss therapy.
Collapse
|
17
|
Münzberg H, Floyd E, Chang JS. Sympathetic Innervation of White Adipose Tissue: to Beige or Not to Beige? Physiology (Bethesda) 2021; 36:246-255. [PMID: 34159808 DOI: 10.1152/physiol.00038.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Obesity research progresses in understanding neuronal circuits and adipocyte biology to regulate metabolism. However, the interface of neuro-adipocyte interaction is less studied. We summarize the current knowledge of adipose tissue innervation and interaction with adipocytes and emphasize adipocyte transitions from white to brown adipocytes and vice versa. We further highlight emerging concepts for the differential neuronal regulation of brown/beige versus white adipocyte and the interdependence of both for metabolic regulation.
Collapse
Affiliation(s)
- Heike Münzberg
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Elizabeth Floyd
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Ji Suk Chang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| |
Collapse
|
18
|
Castorena CM, Caron A, Michael NJ, Ahmed NI, Arnold AG, Lee J, Lee C, Limboy C, Tinajero AS, Granier M, Wang S, Horton JD, Holland WL, Lee S, Liu C, Fujikawa T, Elmquist JK. CB1Rs in VMH neurons regulate glucose homeostasis but not body weight. Am J Physiol Endocrinol Metab 2021; 321:E146-E155. [PMID: 34097543 PMCID: PMC8321828 DOI: 10.1152/ajpendo.00044.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cannabinoid 1 receptor (CB1R) inverse agonists reduce body weight and improve several parameters of glucose homeostasis. However, these drugs have also been associated with deleterious side effects. CB1R expression is widespread in the brain and in peripheral tissues, but whether specific sites of expression can mediate the beneficial metabolic effects of CB1R drugs, while avoiding the untoward side effects, remains unclear. Evidence suggests inverse agonists may act on key sites within the central nervous system to improve metabolism. The ventromedial hypothalamus (VMH) is a critical node regulating energy balance and glucose homeostasis. To determine the contributions of CB1Rs expressed in VMH neurons in regulating metabolic homeostasis, we generated mice lacking CB1Rs in the VMH. We found that the deletion of CB1Rs in the VMH did not affect body weight in chow- and high-fat diet-fed male and female mice. We also found that deletion of CB1Rs in the VMH did not alter weight loss responses induced by the CB1R inverse agonist SR141716. However, we did find that CB1Rs of the VMH regulate parameters of glucose homeostasis independent of body weight in diet-induced obese male mice.NEW & NOTEWORTHY Cannabinoid 1 receptors (CB1Rs) regulate metabolic homeostasis, and CB1R inverse agonists reduce body weight and improve parameters of glucose metabolism. However, the cell populations expressing CB1Rs that regulate metabolic homeostasis remain unclear. CB1Rs are highly expressed in the ventromedial hypothalamic nucleus (VMH), which is a crucial node that regulates metabolism. With CRISPR/Cas9, we generated mice lacking CB1Rs specifically in VMH neurons and found that CB1Rs in VMH neurons are essential for the regulation of glucose metabolism independent of body weight regulation.
Collapse
Affiliation(s)
- Carlos M Castorena
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexandre Caron
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Natalie J Michael
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Newaz I Ahmed
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Amanda G Arnold
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jiwon Lee
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Charlotte Lee
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chelsea Limboy
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Arely Salazar Tinajero
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Madison Granier
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Simeng Wang
- Department of Molecular Genetics, University of Utah, Salt Lake City, Utah
| | - Jay D Horton
- Department of Molecular Genetics, University of Utah, Salt Lake City, Utah
| | - William L Holland
- Department of Biochemistry, Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Syann Lee
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chen Liu
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Teppei Fujikawa
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joel K Elmquist
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
19
|
New Insights of SF1 Neurons in Hypothalamic Regulation of Obesity and Diabetes. Int J Mol Sci 2021; 22:ijms22126186. [PMID: 34201257 PMCID: PMC8229730 DOI: 10.3390/ijms22126186] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 12/16/2022] Open
Abstract
Despite the substantial role played by the hypothalamus in the regulation of energy balance and glucose homeostasis, the exact mechanisms and neuronal circuits underlying this regulation remain poorly understood. In the last 15 years, investigations using transgenic models, optogenetic, and chemogenetic approaches have revealed that SF1 neurons in the ventromedial hypothalamus are a specific lead in the brain’s ability to sense glucose levels and conduct insulin and leptin signaling in energy expenditure and glucose homeostasis, with minor feeding control. Deletion of hormonal receptors, nutritional sensors, or synaptic receptors in SF1 neurons triggers metabolic alterations mostly appreciated under high-fat feeding, indicating that SF1 neurons are particularly important for metabolic adaptation in the early stages of obesity. Although these studies have provided exciting insight into the implications of hypothalamic SF1 neurons on whole-body energy homeostasis, new questions have arisen from these results. Particularly, the existence of neuronal sub-populations of SF1 neurons and the intricate neurocircuitry linking these neurons with other nuclei and with the periphery. In this review, we address the most relevant studies carried out in SF1 neurons to date, to provide a global view of the central role played by these neurons in the pathogenesis of obesity and diabetes.
Collapse
|
20
|
Rawlinson S, Andrews ZB. Hypothalamic insulin signalling as a nexus regulating mood and metabolism. J Neuroendocrinol 2021; 33:e12939. [PMID: 33634518 DOI: 10.1111/jne.12939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 01/23/2023]
Abstract
Insulin has long been known as a metabolic hormone critical in the treatment of diabetes for its peripheral effects on blood glucose. However, in the last 50 years, insulin has entered the realm of neuroendocrinology and many studies have described its function on insulin receptors in the brain in relation to both metabolic and mood disorders. Indeed, rodent models of impaired insulin signalling show signs of dysregulated energy and glucose homeostasis, as well as anxiety-like and depressive behaviours. Importantly, many metabolic diseases such as obesity and diabetes increase the risk of developing mood disorders; however, the brain mechanisms underlying the connection between metabolism and mood remain unresolved. We present the current literature on the importance of the insulin receptor with respect to regulating glucose and energy homeostasis and mood-related behaviours. Specifically, we hypothesise that the insulin receptor in the hypothalamus, classically known as the homeostatic centre of the brain, plays a causal role in linking metabolic and behavioural effects of insulin signalling. In this review, we discuss insulin signalling in the hypothalamus as a critical point of neural integration controlling metabolism and mood.
Collapse
Affiliation(s)
- Sasha Rawlinson
- Department of Physiology, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Zane B Andrews
- Department of Physiology, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
An excitatory ventromedial hypothalamus to paraventricular thalamus circuit that suppresses food intake. Nat Commun 2020; 11:6326. [PMID: 33303759 PMCID: PMC7728757 DOI: 10.1038/s41467-020-20093-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
It is well recognized that ventromedial hypothalamus (VMH) serves as a satiety center in the brain. However, the feeding circuit for the VMH regulation of food intake remains to be defined. Here, we combine fiber photometry, chemo/optogenetics, virus-assisted retrograde tracing, ChR2-assisted circuit mapping and behavioral assays to show that selective activation of VMH neurons expressing steroidogenic factor 1 (SF1) rapidly inhibits food intake, VMH SF1 neurons project dense fibers to the paraventricular thalamus (PVT), selective chemo/optogenetic stimulation of the PVT-projecting SF1 neurons or their projections to the PVT inhibits food intake, and chemical genetic inactivation of PVT neurons diminishes SF1 neural inhibition of feeding. We also find that activation of SF1 neurons or their projections to the PVT elicits a flavor aversive effect, and selective optogenetic stimulation of ChR2-expressing SF1 projections to the PVT elicits direct excitatory postsynaptic currents. Together, our data reveal a neural circuit from VMH to PVT that inhibits food intake. The ventromedial hypothalamus (VMH) serves as a satiety center in the brain, however, the neural circuits involved are incompletely understood. Here, the authors decipher a neural circuit from VMH to the paraventricular thalamus that suppresses food intake.
Collapse
|
22
|
The Role of Ventromedial Hypothalamus Receptors in the Central Regulation of Food Intake. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10120-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
23
|
Fagan MP, Ameroso D, Meng A, Rock A, Maguire J, Rios M. Essential and sex-specific effects of mGluR5 in ventromedial hypothalamus regulating estrogen signaling and glucose balance. Proc Natl Acad Sci U S A 2020; 117:19566-19577. [PMID: 32719118 PMCID: PMC7430975 DOI: 10.1073/pnas.2011228117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The ventromedial hypothalamus (VMH) plays chief roles regulating energy and glucose homeostasis and is sexually dimorphic. We discovered that expression of metabotropic glutamate receptor subtype 5 (mGluR5) in the VMH is regulated by caloric status in normal mice and reduced in brain-derived neurotrophic factor (BDNF) mutants, which are severely obese and have diminished glucose balance control. These findings led us to investigate whether mGluR5 might act downstream of BDNF to critically regulate VMH neuronal activity and metabolic function. We found that mGluR5 depletion in VMH SF1 neurons did not affect energy balance regulation. However, it significantly impaired insulin sensitivity, glycemic control, lipid metabolism, and sympathetic output in females but not in males. These sex-specific deficits are linked to reductions in intrinsic excitability and firing rate of SF1 neurons. Abnormal excitatory and inhibitory synapse assembly and elevated expression of the GABAergic synthetic enzyme GAD67 also cooperate to decrease and potentiate the synaptic excitatory and inhibitory tone onto mutant SF1 neurons, respectively. Notably, these alterations arise from disrupted functional interactions of mGluR5 with estrogen receptors that switch the normally positive effects of estrogen on SF1 neuronal activity and glucose balance control to paradoxical and detrimental. The collective data inform an essential central mechanism regulating metabolic function in females and underlying the protective effects of estrogen against metabolic disease.
Collapse
Affiliation(s)
- Micaella P Fagan
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Anna Rock
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| | - Jamie Maguire
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111;
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
24
|
Rivera P, Tovar R, Ramírez-López MT, Navarro JA, Vargas A, Suárez J, de Fonseca FR. Sex-Specific Anxiety and Prefrontal Cortex Glutamatergic Dysregulation Are Long-Term Consequences of Pre-and Postnatal Exposure to Hypercaloric Diet in a Rat Model. Nutrients 2020; 12:nu12061829. [PMID: 32575416 PMCID: PMC7353464 DOI: 10.3390/nu12061829] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Both maternal and early life malnutrition can cause long-term behavioral changes in the offspring, which depends on the caloric availability and the timing of the exposure. Here we investigated in a rat model whether a high-caloric palatable diet given to the mother and/or to the offspring during the perinatal and/or postnatal period might dysregulate emotional behavior and prefrontal cortex function in the offspring at adult age. To this end, we examined both anxiety responses and the mRNA/protein expression of glutamatergic, GABAergic and endocannabinoid signaling pathways in the prefrontal cortex of adult offspring. Male animals born from mothers fed the palatable diet, and who continued with this diet after weaning, exhibited anxiety associated with an overexpression of the mRNA of Grin1, Gria1 and Grm5 glutamate receptors in the prefrontal cortex. In addition, these animals had a reduced expression of the endocannabinoid system, the main inhibitory retrograde input to glutamate synapses, reflected in a decrease of the Cnr1 receptor and the Nape-pld enzyme. In conclusion, a hypercaloric maternal diet induces sex-dependent anxiety, associated with alterations in both glutamatergic and cannabinoid signaling in the prefrontal cortex, which are accentuated with the continuation of the palatable diet during the life of the offspring.
Collapse
Affiliation(s)
- Patricia Rivera
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
- Correspondence: (P.R.); (F.R.d.F.); Tel.: +34-952-614-012 (P.R. & F.R.d.F.)
| | - Rubén Tovar
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - María Teresa Ramírez-López
- Hospital Universitario de Getafe, Servicio de Ginecología y Obstetricia, 28905 Getafe, Spain;
- Departamento de Enfermería, Facultad de Enfermería, Fisioterapia y Podología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juan Antonio Navarro
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - Antonio Vargas
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
- Correspondence: (P.R.); (F.R.d.F.); Tel.: +34-952-614-012 (P.R. & F.R.d.F.)
| |
Collapse
|
25
|
Aslanpour S, Rosin JM, Balakrishnan A, Klenin N, Blot F, Gradwohl G, Schuurmans C, Kurrasch DM. Ascl1 is required to specify a subset of ventromedial hypothalamic neurons. Development 2020; 147:dev180067. [PMID: 32253239 DOI: 10.1242/dev.180067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 03/23/2020] [Indexed: 03/01/2024]
Abstract
Despite clear physiological roles, the ventromedial hypothalamus (VMH) developmental programs are poorly understood. Here, we asked whether the proneural gene achaete-scute homolog 1 (Ascl1) contributes to VMH development. Ascl1 transcripts were detected in embryonic day (E) 10.5 to postnatal day 0 VMH neural progenitors. The elimination of Ascl1 reduced the number of VMH neurons at E12.5 and E15.5, particularly within the VMH-central (VMHC) and -dorsomedial (VMHDM) subdomains, and resulted in a VMH cell fate change from glutamatergic to GABAergic. We observed a loss of Neurog3 expression in Ascl1-/- hypothalamic progenitors and an upregulation of Neurog3 when Ascl1 was overexpressed. We also demonstrated a glutamatergic to GABAergic fate switch in Neurog3-null mutant mice, suggesting that Ascl1 might act via Neurog3 to drive VMH cell fate decisions. We also showed a concomitant increase in expression of the central GABAergic fate determinant Dlx1/2 in the Ascl1-null hypothalamus. However, Ascl1 was not sufficient to induce an ectopic VMH fate when overexpressed outside the normal window of competency. Combined, Ascl1 is required but not sufficient to specify the neurotransmitter identity of VMH neurons, acting in a transcriptional cascade with Neurog3.
Collapse
Affiliation(s)
- Shaghayegh Aslanpour
- Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jessica M Rosin
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Anjali Balakrishnan
- Sunnybrook Research Institute, Department of Biochemistry, University of Toronto, ON M4N 3M5, Canada
| | - Natalia Klenin
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Florence Blot
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, Universite de Strasbourg, Illkirch 67400, France
| | - Gerard Gradwohl
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, Universite de Strasbourg, Illkirch 67400, France
| | - Carol Schuurmans
- Sunnybrook Research Institute, Department of Biochemistry, University of Toronto, ON M4N 3M5, Canada
| | - Deborah M Kurrasch
- Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
26
|
Glendining KA, Fisher LC, Jasoni CL. Maternal Obesity Modulates Expression of Satb2 in Hypothalamic VMN of Female Offspring. Life (Basel) 2020; 10:life10040048. [PMID: 32344561 PMCID: PMC7235991 DOI: 10.3390/life10040048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 01/24/2023] Open
Abstract
Maternal obesity during pregnancy is associated with a greater risk of poor health outcomes in offspring, including obesity, metabolic disorders, and anxiety, however the incidence of these diseases differs for males and females. Similarly, animal models of maternal obesity have reported sex differences in offspring, for both metabolic outcomes and anxiety-like behaviors. The ventromedial nucleus of the hypothalamus (VMN) is a brain region known to be involved in the regulation of both metabolism and anxiety, and is well documented to be sexually dimorphic. As the VMN is largely composed of glutamatergic neurons, which are important for its functions in modulating metabolism and anxiety, we hypothesized that maternal obesity may alter the number of glutamatergic neurons in the offspring VMN. We used a mouse model of a maternal high-fat diet (mHFD), to examine mRNA expression of the glutamatergic neuronal marker Satb2 in the mediobasal hypothalamus of control and mHFD offspring at GD17.5. We found sex differences in Satb2 expression, with mHFD-induced upregulation of Satb2 mRNA in the mediobasal hypothalamus of female offspring, compared to controls, but not males. Using immunohistochemistry, we found an increase in the number of SATB2-positive cells in female mHFD offspring VMN, compared to controls, which was localized to the rostral region of the nucleus. These data provide evidence that maternal nutrition during gestation alters the developing VMN, possibly increasing its glutamatergic drive of offspring in a sex-specific manner, which may contribute to sexual dimorphism in offspring health outcomes later in life.
Collapse
|
27
|
Neurog2 Acts as a Classical Proneural Gene in the Ventromedial Hypothalamus and Is Required for the Early Phase of Neurogenesis. J Neurosci 2020; 40:3549-3563. [PMID: 32273485 DOI: 10.1523/jneurosci.2610-19.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/28/2022] Open
Abstract
The tuberal hypothalamus is comprised of the dorsomedial, ventromedial, and arcuate nuclei, as well as parts of the lateral hypothalamic area, and it governs a wide range of physiologies. During neurogenesis, tuberal hypothalamic neurons are thought to be born in a dorsal-to-ventral and outside-in pattern, although the accuracy of this description has been questioned over the years. Moreover, the intrinsic factors that control the timing of neurogenesis in this region are poorly characterized. Proneural genes, including Achate-scute-like 1 (Ascl1) and Neurogenin 3 (Neurog3) are widely expressed in hypothalamic progenitors and contribute to lineage commitment and subtype-specific neuronal identifies, but the potential role of Neurogenin 2 (Neurog2) remains unexplored. Birthdating in male and female mice showed that tuberal hypothalamic neurogenesis begins as early as E9.5 in the lateral hypothalamic and arcuate and rapidly expands to dorsomedial and ventromedial neurons by E10.5, peaking throughout the region by E11.5. We confirmed an outside-in trend, except for neurons born at E9.5, and uncovered a rostrocaudal progression but did not confirm a dorsal-ventral patterning to tuberal hypothalamic neuronal birth. In the absence of Neurog2, neurogenesis stalls, with a significant reduction in early-born BrdU+ cells but no change at later time points. Further, the loss of Ascl1 yielded a similar delay in neuronal birth, suggesting that Ascl1 cannot rescue the loss of Neurog2 and that these proneural genes act independently in the tuberal hypothalamus. Together, our findings show that Neurog2 functions as a classical proneural gene to regulate the temporal progression of tuberal hypothalamic neurogenesis.SIGNIFICANCE STATEMENT Here, we investigated the general timing and pattern of neurogenesis within the tuberal hypothalamus. Our results confirmed an outside-in trend of neurogenesis and uncovered a rostrocaudal progression. We also showed that Neurog2 acts as a classical proneural gene and is responsible for regulating the birth of early-born neurons within the ventromedial hypothalamus, acting independently of Ascl1 In addition, we revealed a role for Neurog2 in cell fate specification and differentiation of ventromedial -specific neurons. Last, Neurog2 does not have cross-inhibitory effects on Neurog1, Neurog3, and Ascl1 These findings are the first to reveal a role for Neurog2 in hypothalamic development.
Collapse
|
28
|
Velasco ER, Florido A, Milad MR, Andero R. Sex differences in fear extinction. Neurosci Biobehav Rev 2019; 103:81-108. [PMID: 31129235 PMCID: PMC6692252 DOI: 10.1016/j.neubiorev.2019.05.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/08/2019] [Accepted: 05/19/2019] [Indexed: 12/18/2022]
Abstract
Despite the exponential increase in fear research during the last years, few studies have included female subjects in their design. The need to include females arises from the knowledge gap of mechanistic processes underlying the behavioral and neural differences observed in fear extinction. Moreover, the exact contribution of sex and hormones in relation to learning and behavior is still largely unknown. Insights from this field could be beneficial as fear-related disorders are twice as prevalent in women compared to men. Here, we review an up-to-date summary of animal and human studies in adulthood that report sex differences in fear extinction from a structural and functional approach. Furthermore, we describe how these factors could contribute to the observed sex differences in fear extinction during normal and pathological conditions.
Collapse
Affiliation(s)
- E R Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - A Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - M R Milad
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - R Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
29
|
Neurobiological characteristics underlying metabolic differences between males and females. Prog Neurobiol 2018; 176:18-32. [PMID: 30194984 DOI: 10.1016/j.pneurobio.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/22/2018] [Accepted: 09/01/2018] [Indexed: 12/24/2022]
Abstract
The hypothalamus is the main integrating center for metabolic control. Our understanding of how hypothalamic circuits function to control appetite and energy expenditure has increased dramatically in recent years, due to the rapid rise in the incidence of obesity and the search for effective treatments. Increasing evidence indicates that these treatments will most likely differ between males and females. Indeed, sex differences in metabolism have been demonstrated at various levels, including in two of the most studied neuronal populations involved in metabolic control: the anorexigenic proopiomelanocortin neurons and the orexigenic neuropeptide Y/Agouti-related protein neurons. Here we review what is known to date regarding the sex differences in these two neuronal populations, as well as other neuronal populations involved in metabolic control and glial cells.
Collapse
|
30
|
Viskaitis P, Irvine EE, Smith MA, Choudhury AI, Alvarez-Curto E, Glegola JA, Hardy DG, Pedroni SMA, Paiva Pessoa MR, Fernando ABP, Katsouri L, Sardini A, Ungless MA, Milligan G, Withers DJ. Modulation of SF1 Neuron Activity Coordinately Regulates Both Feeding Behavior and Associated Emotional States. Cell Rep 2018; 21:3559-3572. [PMID: 29262334 PMCID: PMC5746599 DOI: 10.1016/j.celrep.2017.11.089] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/18/2017] [Accepted: 11/27/2017] [Indexed: 12/24/2022] Open
Abstract
Feeding requires the integration of homeostatic drives with emotional states relevant to food procurement in potentially hostile environments. The ventromedial hypothalamus (VMH) regulates feeding and anxiety, but how these are controlled in a concerted manner remains unclear. Using pharmacogenetic, optogenetic, and calcium imaging approaches with a battery of behavioral assays, we demonstrate that VMH steroidogenic factor 1 (SF1) neurons constitute a nutritionally sensitive switch, modulating the competing motivations of feeding and avoidance of potentially dangerous environments. Acute alteration of SF1 neuronal activity alters food intake via changes in appetite and feeding-related behaviors, including locomotion, exploration, anxiety, and valence. In turn, intrinsic SF1 neuron activity is low during feeding and increases with both feeding termination and stress. Our findings identify SF1 neurons as a key part of the neurocircuitry that controls both feeding and related affective states, giving potential insights into the relationship between disordered eating and stress-associated psychological disorders in humans.
Collapse
Affiliation(s)
- Paulius Viskaitis
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Elaine E Irvine
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Mark A Smith
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Agharul I Choudhury
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Elisa Alvarez-Curto
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Justyna A Glegola
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Darran G Hardy
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Silvia M A Pedroni
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Maria R Paiva Pessoa
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Anushka B P Fernando
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Loukia Katsouri
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Alessandro Sardini
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Mark A Ungless
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Dominic J Withers
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
31
|
Eachus H, Bright C, Cunliffe VT, Placzek M, Wood JD, Watt PJ. Disrupted-in-Schizophrenia-1 is essential for normal hypothalamic-pituitary-interrenal (HPI) axis function. Hum Mol Genet 2017; 26:1992-2005. [PMID: 28334933 PMCID: PMC5437527 DOI: 10.1093/hmg/ddx076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/23/2017] [Indexed: 02/01/2023] Open
Abstract
Psychiatric disorders arise due to an interplay of genetic and environmental factors, including stress. Studies in rodents have shown that mutants for Disrupted-In-Schizophrenia-1 (DISC1), a well-accepted genetic risk factor for mental illness, display abnormal behaviours in response to stress, but the mechanisms through which DISC1 affects stress responses remain poorly understood. Using two lines of zebrafish homozygous mutant for disc1, we investigated behaviour and functioning of the hypothalamic-pituitary-interrenal (HPI) axis, the fish equivalent of the hypothalamic-pituitary-adrenal (HPA) axis. Here, we show that the role of DISC1 in stress responses is evolutionarily conserved and that DISC1 is essential for normal functioning of the HPI axis. Adult zebrafish homozygous mutant for disc1 show aberrant behavioural responses to stress. Our studies reveal that in the embryo, disc1 is expressed in neural progenitor cells of the hypothalamus, a conserved region of the vertebrate brain that centrally controls responses to environmental stressors. In disc1 mutant embryos, proliferating rx3+ hypothalamic progenitors are not maintained normally and neuronal differentiation is compromised: rx3-derived ff1b+ neurons, implicated in anxiety-related behaviours, and corticotrophin releasing hormone (crh) neurons, key regulators of the stress axis, develop abnormally, and rx3-derived pomc+ neurons are disorganised. Abnormal hypothalamic development is associated with dysfunctional behavioural and neuroendocrine stress responses. In contrast to wild type siblings, disc1 mutant larvae show altered crh levels, fail to upregulate cortisol levels when under stress and do not modulate shoal cohesion, indicative of abnormal social behaviour. These data indicate that disc1 is essential for normal development of the hypothalamus and for the correct functioning of the HPA/HPI axis.
Collapse
Affiliation(s)
- Helen Eachus
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.,The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Charlotte Bright
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Vincent T Cunliffe
- The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Marysia Placzek
- The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Jonathan D Wood
- The Bateson Centre, Department of Biomedical Science, Firth Court, Western Bank, Sheffield S10 2TN, UK.,Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Penelope J Watt
- Department of Animal and Plant Sciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
32
|
Yang T, Yang CF, Chizari MD, Maheswaranathan N, Burke KJ, Borius M, Inoue S, Chiang MC, Bender KJ, Ganguli S, Shah NM. Social Control of Hypothalamus-Mediated Male Aggression. Neuron 2017; 95:955-970.e4. [PMID: 28757304 PMCID: PMC5648542 DOI: 10.1016/j.neuron.2017.06.046] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/13/2017] [Accepted: 06/27/2017] [Indexed: 12/23/2022]
Abstract
How environmental and physiological signals interact to influence neural circuits underlying developmentally programmed social interactions such as male territorial aggression is poorly understood. We have tested the influence of sensory cues, social context, and sex hormones on progesterone receptor (PR)-expressing neurons in the ventromedial hypothalamus (VMH) that are critical for male territorial aggression. We find that these neurons can drive aggressive displays in solitary males independent of pheromonal input, gonadal hormones, opponents, or social context. By contrast, these neurons cannot elicit aggression in socially housed males that intrude in another male's territory unless their pheromone-sensing is disabled. This modulation of aggression cannot be accounted for by linear integration of environmental and physiological signals. Together, our studies suggest that fundamentally non-linear computations enable social context to exert a dominant influence on developmentally hard-wired hypothalamus-mediated male territorial aggression.
Collapse
Affiliation(s)
- Taehong Yang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Cindy F Yang
- Program in Neuroscience, UC San Francisco, San Francisco, CA 94158, USA
| | - M Delara Chizari
- Department of Anatomy, UC San Francisco, San Francisco, CA 94158, USA
| | | | - Kenneth J Burke
- Program in Neuroscience, UC San Francisco, San Francisco, CA 94158, USA
| | - Maxim Borius
- Department of Anatomy, UC San Francisco, San Francisco, CA 94158, USA
| | - Sayaka Inoue
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Michael C Chiang
- Department of Anatomy, UC San Francisco, San Francisco, CA 94158, USA
| | - Kevin J Bender
- Department of Neurology, UC San Francisco, San Francisco, CA 94158, USA
| | - Surya Ganguli
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Nirao M Shah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Martínez-Sánchez N, Seoane-Collazo P, Contreras C, Varela L, Villarroya J, Rial-Pensado E, Buqué X, Aurrekoetxea I, Delgado TC, Vázquez-Martínez R, González-García I, Roa J, Whittle AJ, Gomez-Santos B, Velagapudi V, Tung YCL, Morgan DA, Voshol PJ, Martínez de Morentin PB, López-González T, Liñares-Pose L, Gonzalez F, Chatterjee K, Sobrino T, Medina-Gómez G, Davis RJ, Casals N, Orešič M, Coll AP, Vidal-Puig A, Mittag J, Tena-Sempere M, Malagón MM, Diéguez C, Martínez-Chantar ML, Aspichueta P, Rahmouni K, Nogueiras R, Sabio G, Villarroya F, López M. Hypothalamic AMPK-ER Stress-JNK1 Axis Mediates the Central Actions of Thyroid Hormones on Energy Balance. Cell Metab 2017; 26:212-229.e12. [PMID: 28683288 PMCID: PMC5501726 DOI: 10.1016/j.cmet.2017.06.014] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/17/2017] [Accepted: 06/15/2017] [Indexed: 02/02/2023]
Abstract
Thyroid hormones (THs) act in the brain to modulate energy balance. We show that central triiodothyronine (T3) regulates de novo lipogenesis in liver and lipid oxidation in brown adipose tissue (BAT) through the parasympathetic (PSNS) and sympathetic nervous system (SNS), respectively. Central T3 promotes hepatic lipogenesis with parallel stimulation of the thermogenic program in BAT. The action of T3 depends on AMP-activated protein kinase (AMPK)-induced regulation of two signaling pathways in the ventromedial nucleus of the hypothalamus (VMH): decreased ceramide-induced endoplasmic reticulum (ER) stress, which promotes BAT thermogenesis, and increased c-Jun N-terminal kinase (JNK) activation, which controls hepatic lipid metabolism. Of note, ablation of AMPKα1 in steroidogenic factor 1 (SF1) neurons of the VMH fully recapitulated the effect of central T3, pointing to this population in mediating the effect of central THs on metabolism. Overall, these findings uncover the underlying pathways through which central T3 modulates peripheral metabolism.
Collapse
Affiliation(s)
- Noelia Martínez-Sánchez
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Patricia Seoane-Collazo
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Cristina Contreras
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Luis Varela
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Joan Villarroya
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona (IBUB), Barcelona 08028, Spain; Hospital de la Santa Creu i Sant Pau, Barcelona 08026, Spain
| | - Eva Rial-Pensado
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Xabier Buqué
- Department of Physiology, University of the Basque Country UPV/EHU, Biocruces Research Institute, Barakaldo 48903, Spain
| | - Igor Aurrekoetxea
- Department of Physiology, University of the Basque Country UPV/EHU, Biocruces Research Institute, Barakaldo 48903, Spain
| | - Teresa C Delgado
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Derio, Bizkaia 48160, Spain
| | - Rafael Vázquez-Martínez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | - Ismael González-García
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Juan Roa
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | - Andrew J Whittle
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Beatriz Gomez-Santos
- Department of Physiology, University of the Basque Country UPV/EHU, Biocruces Research Institute, Barakaldo 48903, Spain
| | - Vidya Velagapudi
- VTT Technical Research Centre of Finland, Tietotie 2, Espoo FIN-02044, Finland; Metabolomics Unit, Institute for Molecular Medicine, University of Helsinki, Helsinki FI-00290, Finland
| | - Y C Loraine Tung
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Peter J Voshol
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Pablo B Martínez de Morentin
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Tania López-González
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Clinical Neurosciences Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Laura Liñares-Pose
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Francisco Gonzalez
- Department of Surgery, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Service of Ophthalmology, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela 15706, Spain
| | - Krishna Chatterjee
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Gema Medina-Gómez
- University Rey Juan Carlos, Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Avda. de Atenas s/n, Alcorcon, Madrid 28922, Spain
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA
| | - Núria Casals
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallés, Barcelona 08195, Spain
| | - Matej Orešič
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku FI-20520, Finland
| | - Anthony P Coll
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Jens Mittag
- University of Lübeck, Internal Medicine I, Center of Brain, Behavior, and Metabolism (CBBM), Ratzeburger Allee 160, Lübeck 23562, Germany
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba 14004, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, Turku FIN-20520, Finland
| | - María M Malagón
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - María Luz Martínez-Chantar
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Derio, Bizkaia 48160, Spain
| | - Patricia Aspichueta
- Department of Physiology, University of the Basque Country UPV/EHU, Biocruces Research Institute, Barakaldo 48903, Spain
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Rubén Nogueiras
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Guadalupe Sabio
- Myocardial Pathophysiology, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Francesc Villarroya
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona (IBUB), Barcelona 08028, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| |
Collapse
|
34
|
Xu Y, O'Malley BW, Elmquist JK. Brain nuclear receptors and body weight regulation. J Clin Invest 2017; 127:1172-1180. [PMID: 28218618 DOI: 10.1172/jci88891] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neural pathways, especially those in the hypothalamus, integrate multiple nutritional, hormonal, and neural signals, resulting in the coordinated control of body weight balance and glucose homeostasis. Nuclear receptors (NRs) sense changing levels of nutrients and hormones, and therefore play essential roles in the regulation of energy homeostasis. Understanding the role and the underlying mechanisms of NRs in the context of energy balance control may facilitate the identification of novel targets to treat obesity. Notably, NRs are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of energy balance, including feeding, energy expenditure and physical activity. In this Review we summarize some of the recent literature regarding effects of brain NRs on body weight regulation and discuss mechanisms underlying these effects.
Collapse
|
35
|
Origins and Functions of the Ventrolateral VMH: A Complex Neuronal Cluster Orchestrating Sex Differences in Metabolism and Behavior. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:199-213. [PMID: 29224096 DOI: 10.1007/978-3-319-70178-3_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The neuroendocrine brain or hypothalamus has emerged as one of the most highly sexually dimorphic brain regions in mammals, and specifically in rodents. It is not surprising that hypothalamic nuclei play a pivotal role in controlling sex-dependent physiology. This brain region functions as a chief executive officer or master regulator of homeostatic physiological systems to integrate both external and internal signals. In this review, we describe sex differences in energy homeostasis that arise in one area of the hypothalamus, the ventrolateral subregion of the ventromedial hypothalamus (VMHvl) with a focus on how male and female neurons function in metabolic and behavioral aspects. Because other chapters within this book provide details on signaling pathways in the VMH that contribute to sex differences in metabolism, our discussion will be limited to how the sexually dimorphic VMHvl develops and what key regulators are thought to control the many functional and physiological endpoints attributed to this region. In the last decade, several exciting new studies using state-of-the-art genetic and molecular tools are beginning to provide some understanding as to how specific neurons contribute to the coordinated physiological responses needed by male and females. New technology that combines intersectional spatial and genetic approaches is now allowing further refinement in how we describe, probe, and manipulate critical male and female neurocircuits involved in metabolism.
Collapse
|
36
|
Silva BA, Gross CT, Gräff J. The neural circuits of innate fear: detection, integration, action, and memorization. ACTA ACUST UNITED AC 2016; 23:544-55. [PMID: 27634145 PMCID: PMC5026211 DOI: 10.1101/lm.042812.116] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/19/2016] [Indexed: 12/26/2022]
Abstract
How fear is represented in the brain has generated a lot of research attention, not only because fear increases the chances for survival when appropriately expressed but also because it can lead to anxiety and stress-related disorders when inadequately processed. In this review, we summarize recent progress in the understanding of the neural circuits processing innate fear in rodents. We propose that these circuits are contained within three main functional units in the brain: a detection unit, responsible for gathering sensory information signaling the presence of a threat; an integration unit, responsible for incorporating the various sensory information and recruiting downstream effectors; and an output unit, in charge of initiating appropriate bodily and behavioral responses to the threatful stimulus. In parallel, the experience of innate fear also instructs a learning process leading to the memorization of the fearful event. Interestingly, while the detection, integration, and output units processing acute fear responses to different threats tend to be harbored in distinct brain circuits, memory encoding of these threats seems to rely on a shared learning system.
Collapse
Affiliation(s)
- Bianca A Silva
- Laboratory of Neuroepigenetics, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale Lausanne, CH-1015 Lausanne, Switzerland
| | - Cornelius T Gross
- Mouse Biology Unit, European Molecular Biology Laboratory (EMBL), 00015 Monterotondo, Italy
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
37
|
Sohn JW, Oh Y, Kim KW, Lee S, Williams KW, Elmquist JK. Leptin and insulin engage specific PI3K subunits in hypothalamic SF1 neurons. Mol Metab 2016; 5:669-679. [PMID: 27656404 PMCID: PMC5021675 DOI: 10.1016/j.molmet.2016.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 11/21/2022] Open
Abstract
Objective The ventromedial hypothalamic nucleus (VMH) regulates energy balance and glucose homeostasis. Leptin and insulin exert metabolic effects via their cognate receptors expressed by the steroidogenic factor 1 (SF1) neurons within the VMH. However, detailed cellular mechanisms involved in the regulation of these neurons by leptin and insulin remain to be identified. Methods We utilized genetically-modified mouse models and performed patch-clamp electrophysiology experiments to resolve this issue. Results We identified distinct populations of leptin-activated and leptin-inhibited SF1 neurons. In contrast, insulin uniformly inhibited SF1 neurons. Notably, we found that leptin-activated, leptin-inhibited, and insulin-inhibited SF1 neurons are distinct subpopulations within the VMH. Leptin depolarization of SF1 neuron also required the PI3K p110β catalytic subunit. This effect was mediated by the putative transient receptor potential C (TRPC) channel. On the other hand, hyperpolarizing responses of SF1 neurons by leptin and insulin required either of the p110α or p110β catalytic subunits, and were mediated by the putative ATP-sensitive K+ (KATP) channel. Conclusions Our results demonstrate that specific PI3K catalytic subunits are responsible for the acute effects of leptin and insulin on VMH SF1 neurons, and provide insights into the cellular mechanisms of leptin and insulin action on VMH SF1 neurons that regulate energy balance and glucose homeostasis. Leptin recruits p110β/TRPC channels to depolarize/activate SF1 neurons. Leptin recruits p110α/p110β/KATP channels to hyperpolarize/inhibit SF1 neurons. Insulin recruits p110α/p110β/KATP channels to hyperpolarize/inhibit SF1 neurons. Acute leptin and insulin responses are segregated to distinct subsets of VMH SF1 neurons.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea.
| | - Youjin Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Ki Woo Kim
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Pharmacology, Yonsei University Wonju College of Medicine, Wonju, 26426, South Korea
| | - Syann Lee
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kevin W Williams
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Joel K Elmquist
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
38
|
Bayless DW, Shah NM. Genetic dissection of neural circuits underlying sexually dimorphic social behaviours. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150109. [PMID: 26833830 DOI: 10.1098/rstb.2015.0109] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 11/12/2022] Open
Abstract
The unique hormonal, genetic and epigenetic environments of males and females during development and adulthood shape the neural circuitry of the brain. These differences in neural circuitry result in sex-typical displays of social behaviours such as mating and aggression. Like other neural circuits, those underlying sex-typical social behaviours weave through complex brain regions that control a variety of diverse behaviours. For this reason, the functional dissection of neural circuits underlying sex-typical social behaviours has proved to be difficult. However, molecularly discrete neuronal subpopulations can be identified in the heterogeneous brain regions that control sex-typical social behaviours. In addition, the actions of oestrogens and androgens produce sex differences in gene expression within these brain regions, thereby highlighting the neuronal subpopulations most likely to control sexually dimorphic social behaviours. These conditions permit the implementation of innovative genetic approaches that, in mammals, are most highly advanced in the laboratory mouse. Such approaches have greatly advanced our understanding of the functional significance of sexually dimorphic neural circuits in the brain. In this review, we discuss the neural circuitry of sex-typical social behaviours in mice while highlighting the genetic technical innovations that have advanced the field.
Collapse
Affiliation(s)
- Daniel W Bayless
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Nirao M Shah
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|