1
|
Licker M, Ellenberger C. Impact of the Circadian Rhythm and Seasonal Changes on the Outcome of Cardiovascular Interventions. J Clin Med 2025; 14:2570. [PMID: 40283400 PMCID: PMC12028144 DOI: 10.3390/jcm14082570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
The activities of living beings fluctuate according to seasonal changes and circadian rhythms. The interaction of organisms with their environment, notably weather conditions and night-day cycles, modulate homeostatic mechanisms and influence physiological responses in stressful situations. In humans, it is well established that cardiovascular events such as myocardial infarction, stroke and acute heart failure more frequently occur in winter than in summer season (non-tropical regions) and in the morning than in the evening. While the effects of cardiovascular medications vary during the day, the influence of circadian rhythms on the outcomes of invasive interventions is the subject of conflicting debates. This paper analyzes the impact of seasonal variability and circadian rhythms on physiological responses and the occurrence of complications in cardiac surgery and interventional cardiology.
Collapse
Affiliation(s)
- Marc Licker
- Department of Cardiovascular & Thoracic Anaesthesia and Critical Care, University Hospital of Martinique, F-97200 Fort de France, France;
- Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christoph Ellenberger
- Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Department of Acute Medicine, University Hospital of Geneva, CH-1205 Geneva, Switzerland
| |
Collapse
|
2
|
Morales L, Desfilis E, Medina L. Development of catecholaminergic neurons of Otp-lineage in the medial extended amygdala and related forebrain centers. Front Neuroanat 2025; 19:1553952. [PMID: 40177299 PMCID: PMC11961924 DOI: 10.3389/fnana.2025.1553952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Catecholaminergic (CA) neurons of the medial extended amygdala, preoptic region and adjacent alar hypothalamus have been involved in different aspects of social behavior, as well as in modulation of homeostasis in response to different stressors. Previous data suggested that at least some CA neurons of the medial extended amygdala could originate in a hypothalamic embryonic domain that expresses the transcription factor Otp. To investigate this, we used Otp-eGFP mice (with permanent labeling of GFP in Otp cells) to analyze coexpression of GFP and tyrosine hydroxylase (TH) throughout ontogenesis by way of double immunofluorescence. Our results provide evidence that some forebrain CA cells belong to the Otp lineage. In particular, we found small subpopulations of TH cells that coexpress GFP within the medial extended amygdala, the periventricular preoptic area, the paraventricular hypothalamus, the periventricular hypothalamus, as well as some subdivisions of the basal hypothalamus. In some of the Otp cells, such as those of extended amygdala, the expression of TH appears to be transitory, in agreement with previous studies. The results open interesting questions about the role of these Otp versus non-Otp catecholaminergic subpopulations during development, network integration and in modulation of different functions, including homeostasis and social behaviors.
Collapse
Affiliation(s)
- Lorena Morales
- Department of Medicina Experimental, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Ester Desfilis
- Department of Medicina Experimental, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Loreta Medina
- Department of Medicina Experimental, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Spain
| |
Collapse
|
3
|
Wang B, Yu Y, Li J, Xiong Y, Zhang X, Wan Y, Zheng R, Zhang C. Hypothalamic GABAergic neurons: their roles in health and metabolic diseases. Front Endocrinol (Lausanne) 2025; 16:1551741. [PMID: 40130157 PMCID: PMC11930815 DOI: 10.3389/fendo.2025.1551741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
Hypothalamic GABAergic neurons are important in regulating metabolic homeostasis and energy balance. Serving as critical integrators of catabolic and anabolic processes, these neurons orchestrate a broad spectrum of metabolic functions, including feeding, nutrient metabolism, fluid homeostasis, basal metabolism, thermoregulation, and circadian rhythms. Recent advances in neuroscience have facilitated a deeper exploration of the role of hypothalamic GABAergic neurons in metabolic regulation. Emerging research has uncovered key mechanisms through which these neurons modulate energy balance and maintain metabolic balance. These findings not only enhance our understanding of obesity and related metabolic disorders but also underscore the link between hypothalamic dysfunction and prevalent metabolic diseases such as obesity and type 2 diabetes. This review summarizes the latest advancements in our understanding of the role of hypothalamic GABAergic neurons in metabolic regulation. It aims to elucidate the neural and molecular mechanisms underlying hypothalamic control of metabolism, offering new perspectives for the diagnosis and treatment of metabolic disorders.
Collapse
Affiliation(s)
- Bingwei Wang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Nucleic Acid Medicine, Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Yu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Nucleic Acid Medicine, Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Juan Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Xiong
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Nucleic Acid Medicine, Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| | - Chunxiang Zhang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Nucleic Acid Medicine, Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Yan M, Lv X, Zhang S, Song Z, Hu B, Qing X, Kou H, Chen S, Shao Z, Liu H. Alleviation of inflammation in paraventricular nucleus and sympathetic outflow by melatonin efficiently repairs endplate porosities and attenuates spinal hyperalgesia. Int Immunopharmacol 2025; 149:114213. [PMID: 39914282 DOI: 10.1016/j.intimp.2025.114213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/22/2025]
Abstract
Low back pain, largely attributed to intervertebral disc (IVD) degeneration, is correlated with increased sympathetic nerve activity. Toll-like receptor 4 (TLR4)-mediated inflammation in the paraventricular nucleus (PVN) triggers sympathetic nerve activation, which remains uncharted in IVD degeneration. We hypothesized that lumbar spine instability (LSI) surgery in mice elevated sympathetic outflow by activating TLR4/NF-κB axis in PVN, and exacerbated endplate porosities and spinal hyperalgesia following 4 or 8 weeks LSI surgery. Treatment of melatonin for 8 weeks notably alleviated the inflammation and sympathetic outflow in the PVN, and attenuated sympathetic nerve activity, oxidative stress, endplate porosities and spinal hyperalgesia in the peripheral. These effects were abolished by melatonin receptor antagonist luzindole. Immunofluorescent staining of melatonin receptor 1A (MT1) and 1B (MT2) confirmed that MT2 expression exceeded that of MT1 in PVN. Knockdown of MT2 in PVN blocked the inhibitory effect of melatonin on inflammation and sympathetic activation both in PVN and endplate, as well as spinal hyperalgesia, oxidative stress, and porosities of endplate. Additionally, norepinephrine induces inflammation and oxidative stress, disrupts metabolic homeostasis of endplate cells via α2-adrenergic receptor in vitro. This study suggests that melatonin, via activation of MT2, inhibits inflammation and sympathetic activities both in PVN and endplate, therefore, efficiently repairing endplate porosities and alleviating spinal hyperalgesia induced by LSI.
Collapse
Affiliation(s)
- Miaoheng Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Shuo Zhang
- School of Medicine, Nankai University, Tianjin 300071 China
| | - Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 China
| | - Binwu Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Xiangcheng Qing
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Hongwei Kou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 China.
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 China.
| |
Collapse
|
5
|
Ma CM, Zhang FS, Zhao XH, Yang Y, Wang B, Wang Y, Liu XF, Bian X, Xu ZX, Zhang G, Qu LZ, Zhang N. Genistein improves depression-like behavior in rats by regulating intestinal flora and altering glutamate gene expression. Curr Res Food Sci 2025; 10:101020. [PMID: 40124394 PMCID: PMC11930109 DOI: 10.1016/j.crfs.2025.101020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025] Open
Abstract
Depression is a mental disorder, and genistein is known to have antidepressant effects, but its mechanism of action is still unclear. Here, the mechanism of genistein improving depression based on gut microbiota was explored using classic behavioral indicators of depression combined with genomic technology. The behavioral evaluation showed that rats gavaged with 20-40 mg/kg genistein showed an increase in body weight, glucose preference, absenteeism score, body temperature, and 5-hydroxytryptamine (5-HT) content, while a decrease in adrenocorticotropic hormone (ACTH) and corticosterone (CORT) content compared to the depression rat model group, but there was no significant difference compared to the positive control (fluoxetine). The results of high-throughput sequencing showed that genistein increased the relative abundance of Firmicutes and Actinobacteriota and decreased the relative abundance of Bacteroidota at the phylum level. At the genus level, the abundance of Bifidobacterium, a short-chain fatty acid producing bacterium, was increased. Furthermore, metagenome results revealed that the antidepressant effect of genistein can be achieved by promoting glutamate metabolism, increasing glutamic acid decarboxylase (GAD) expression levels, promoting γ-aminobutyric acid (GABA) synthesis, and indirectly increasing 5-HT levels.
Collapse
Affiliation(s)
- Chun-min Ma
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Fu-shun Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Xin-huai Zhao
- School of Biological and Food Engineering, Guangdong University of Petrochemical Technology, 525000, Maoming, PR China
| | - Yang Yang
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Bing Wang
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Yan Wang
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Xiao-fei Liu
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Xin Bian
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Zi-Xuan Xu
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Guang Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| | - Li-zhe Qu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Na Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, PR China
| |
Collapse
|
6
|
He W, Shi L, Yue Z, Zhao K, Wang X, Wang K, Jing X, Bi S, Deng T, Zhao X, Tian X, Ma X, Chen Y, Yuan F, Wang S. Activation of glutamatergic neurons in the organum vasculosum of the lamina terminalis induces thirst-driven sniffing. Cell Rep 2025; 44:115254. [PMID: 39893636 DOI: 10.1016/j.celrep.2025.115254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Sniffing is a specialized respiratory behavior that enables rodents to localize and track objects in their environment. The organum vasculosum of the lamina terminalis (OVLT) is critically involved in the regulation of thirst and water intake, yet its role in controlling thirst-driven exploratory sniffing behaviors remains unclear. This study demonstrates that hypertonic stimulation significantly increases sniffing and activates OVLT glutamatergic (OVLTGlut) neurons. Photostimulation of both OVLTGlut neurons and their axon terminals within the paraventricular nucleus of the hypothalamus (PVN) induces robust sniffing. Furthermore, ablation of PVN neurons projecting to the preBötzinger complex not only reduces the sniffing time induced by photostimulation of OVLTGlut neurons projecting to the PVN but also prolongs the drinking latency. These findings identify the OVLTGlut-PVN-preBötzinger complex circuit as a pivotal regulator of thirst-driven sniffing, providing insights into the neural mechanisms underlying thirst and exploratory behavior.
Collapse
Affiliation(s)
- Wei He
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Luo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Ziteng Yue
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Ke Zhao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoyi Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Kailin Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xinyi Jing
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Shangyu Bi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xue Zhao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaochen Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiangchen Ma
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yongqiang Chen
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.
| |
Collapse
|
7
|
Zhang M, Wang Q, Wang Y. Brain endocannabinoid control of metabolic and non-metabolic feeding behaviors. Neurochem Int 2025; 183:105921. [PMID: 39708909 DOI: 10.1016/j.neuint.2024.105921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The central endocannabinoid (eCB) system in brain shows a crucial role in the regulation of feeding behaviors, influencing both metabolic and non-metabolic mechanisms of appetite control, which has been paid much attention. Although there are already many review articles discussing eCB modulation of feeding behaviors, our paper attempts to summarize the recent advancements through synapses, circuits, and network in brain. Our focus is on the dual role of eCB signalling in regulating metabolic energy balance and hedonic reward-related feeding. In the context of metabolic regulation of feeding behaviors, eCBs affect the hypothalamic circuits that balance hunger and satiety through signal integration related to energy status and nutrient availability. Dysregulation of this system can contribute to metabolic disorders such as obesity and anorexia. In non-metabolic feeding, the eCB system influences the hedonic aspects of eating by modulating reward pathways, including the mesolimbic system and the olfactory bulb, critical for motivating food intake and processing sensory cues. This review also explores therapeutic strategies targeting the eCB system, including cannabinoid receptor antagonists and eCB hydrolase enzyme inhibitors, which hold promise for treating conditions associated with appetite dysregulation and eating disorders. By synthesizing recent findings, we aim to highlight the intricate mechanisms through which the eCB system affects feeding behavior and to propose future directions for research and therapeutic intervention in the realm of appetite control and eating disorders.
Collapse
Affiliation(s)
- Maoxing Zhang
- Basic School of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 260071, China
| | - Qingyu Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ying Wang
- Basic School of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 260071, China.
| |
Collapse
|
8
|
Li B, Du L, Wu S, Yin Y. Transcriptomics study of hippocampus in mice exposed to heat stress. Psychoneuroendocrinology 2025; 172:107279. [PMID: 39798407 DOI: 10.1016/j.psyneuen.2025.107279] [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: 07/09/2024] [Revised: 09/04/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
Heat stress (HS) triggers various pathophysiological responses in the brain, including neuroinflammation and cognitive impairments. The objective of this study was to examine the impact of HS by comparing the hippocampal transcriptomes of mice exposed to HS with those under control conditions. Our analysis revealed that HS exposure did not affect the number of SNP or InDel mutations in the mouse hippocampus, nor did it influence SNP functions, distribution, or types. However, HS did lead to differential gene expression in the hippocampus, with 210 differentially expressed genes (DEGs), including 72 upregulated and 138 downregulated genes. Gene Ontology (GO) analysis indicated that these DEGs are involved in hippocampal responses to various stimuli (chemical, oxygen-containing compounds, peptide hormones), metabolic processes (arachidonic acid, olefinic compound metabolism, lipid metabolism), and other functions. The regulation of these functions may be closely linked to specific DEGs, such as Card14, Ntrk1, Lcn2, Irs4, Cyp2c70, Hamp, Ambp, Gh, Mup19, and others, which exhibit the highest degree of differential variation. Furthermore, we observed that pre-treatment with taurine primarily modulated cognitive functions in the hippocampus following HS. Therefore, our study offers valuable insights for future research on heat stress-induced cognitive impairments and provides a theoretical foundation for developing taurine-based preventive strategies for high-risk populations.
Collapse
Affiliation(s)
- Bin Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
9
|
Lu M, Zhang J, Zhang Q, Sun J, Zou D, Huang J, Liu W. The parasubthalamic nucleus: A novel eating center in the brain. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111250. [PMID: 39788409 DOI: 10.1016/j.pnpbp.2025.111250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/21/2024] [Accepted: 01/04/2025] [Indexed: 01/12/2025]
Abstract
Eating behavior stands as a fundamental determinant of animal survival and growth, intricately regulated by an amalgamation of internal and external stimuli. Coordinated movements of facial muscles and the mandible orchestrate prey capture and food processing, propelled by the allure of taste and rewarding food properties. Conversely, satiation, pain, aversion, negative emotion or perceived threats can precipitate the cessation or avoidance of eating activities. In recent years, the parasubthalamic nucleus (PSTN), located in the lateral hypothalamic area, has emerged as a focal point in feeding research. PSTN neurons assume pivotal roles within multiple feeding circuits, bridging central feeding centers with peripheral organs. They intricately modulate regulation of oral sensorimotor functions, hedonic feeding, appetite motivation and the processing of satiation and aversive signals, thereby orchestrating the initiation or termination of feeding behaviors. This review delves into the distinctive neuronal subpopulations within the PSTN and their associated neural networks, aiming to refine our comprehension of the neural underpinnings of feeding while also seeking to unearth more efficacious therapeutic avenues for feeding and eating disorders.
Collapse
Affiliation(s)
- Mingxuan Lu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Jiayao Zhang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Qi Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China; Shanghai Changning Mental Health Center, Shanghai 200335, China
| | - Jiyu Sun
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Danni Zou
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Jinyin Huang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Weicai Liu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China.
| |
Collapse
|
10
|
Wang T, Teng B, Yao DR, Gao W, Oka Y. Organ-specific sympathetic innervation defines visceral functions. Nature 2025; 637:895-902. [PMID: 39604732 DOI: 10.1038/s41586-024-08269-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
The autonomic nervous system orchestrates the functions of the brain and body through the sympathetic and parasympathetic pathways1. However, our understanding of the autonomic system, especially the sympathetic system, at the cellular and molecular levels is severely limited. Here we show topological representations of individual visceral organs in the major abdominal sympathetic ganglion complex. Using multi-modal transcriptomic analyses, we identified molecularly distinct sympathetic populations in the coeliac-superior mesenteric ganglia (CG-SMG). Of note, individual CG-SMG populations exhibit selective and mutually exclusive axonal projections to visceral organs, targeting either the gastrointestinal tract or secretory areas including the pancreas and bile tract. This combinatorial innervation pattern suggests functional segregation between different CG-SMG populations. Indeed, our neural perturbation experiments demonstrated that one class of neurons regulates gastrointestinal transit, and another class of neurons controls digestion and glucagon secretion independent of gut motility. These results reveal the molecularly diverse sympathetic system and suggest modular regulation of visceral organ functions by sympathetic populations.
Collapse
Affiliation(s)
- Tongtong Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Bochuan Teng
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dickson R Yao
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
11
|
Li B, Du L, Wu S, Yin Y. Protective effects of taurine on heat Stress-Induced cognitive impairment through Npas4 and Lcn2. Int Immunopharmacol 2024; 143:113376. [PMID: 39405930 DOI: 10.1016/j.intimp.2024.113376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
Heat stress (HS) induces various pathophysiological responses in the brain, encompassing neuroinflammation and cognitive impairments. Although taurine has been reported to possess anti-inflammatory and cognitive-enhancing properties, its role and mechanisms in HS-induced cognitive impairment remain unclear. This study supplemented mice exposed to HS with taurine to assess its effect on cognitive function in a HS-induced mouse model. The results revealed that taurine ameliorated cognitive deficits following HS in mice and mitigated HS-induced astrocyte and microglia activation as well as blood-brain barrier (BBB) damage in the hippocampus. Mechanistically, Mechanistically, transcriptome sequencing was employed to identify that taurine regulates neuronal PAS domain protein (Npas4) and lipocalin 2 (Lcn2) during HS. Taurine was found to modulate hippocampal inflammation and influence cognitive function by upregulating Npas4 and downregulating Lcn2 after HS. Subsequently, molecular docking and AnimalTFDB database calculations were conducted, revealing that taurine might regulate the expression of Npas4 and Lcn2 by modulating the regulatory transcription factors (TFs) RE1 silencing transcription factor (REST) and nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1). Our findings demonstrate that taurine enhances the recovery of cognitive function through Npas4 and Lcn2 following HS, providing a theoretical basis for the clinical application of taurine in preventing or treating HS-induced cognitive impairment.
Collapse
Affiliation(s)
- Bin Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
12
|
Zahner MR, Hillard KJ, Chandley MC. The role of the dorsomedial hypothalamus in the cardiogenic sympathetic reflex in the Sprague Dawley rat. Front Physiol 2024; 15:1479892. [PMID: 39777361 PMCID: PMC11703967 DOI: 10.3389/fphys.2024.1479892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
Myocardial ischemia causes the production and release of metabolites such as bradykinin, which stimulates cardiac spinal sensory afferents, causing chest pain and an increase in sympathetic activity referred to as the cardiogenic sympathetic afferent reflex. While the brain stem nuclei, such as the nucleus tractus solitarius and rostral ventrolateral medulla, are essential in the cardiogenic sympathetic afferent reflex, the role of other supramedullary nuclei in the cardiogenic sympathetic afferent reflex are not clear. The dorsomedial hypothalamic nucleus (DMH) is involved in cardiovascular sympathetic regulation and plays an important role in the sympathetic response to stressful stimuli. In this study, we determined the role of DMH in the cardiogenic sympathetic afferent reflex. To do this we measured arterial pressure, heart rate, and renal sympathetic nerve activity (RSNA) responses to epicardial bradykinin (10 μg/mL) in anesthetized Sprague Dawley rats before and after bilateral DMH microinjection (50 nL) of either the GABAA agonist muscimol (0.5 nmol) to inhibit or the antagonist bicuculline (40 pmol) to disinhibit activity. Muscimol inhibition elicited a modest, albeit significant, reduction in basal arterial pressure and heart rate and attenuated the arterial pressure and heart rate reflex response to epicardial bradykinin. However, it did not change the magnitude of the reflex. Bicuculline disinhibition of the DMH increased basal arterial pressure, heart rate, and RSNA but did not augment the response to epicardial bradykinin. These results suggest that sympathetic activity derived from the DMH does not play an important role in the cardiogenic sympathetic afferent reflex in Sprague Dawley rats.
Collapse
Affiliation(s)
- Matthew R. Zahner
- Health Sciences Department, College of Public Health, East Tennessee State University, Johnson City, TN, United States
| | - Kynlee J. Hillard
- Health Sciences Department, College of Public Health, East Tennessee State University, Johnson City, TN, United States
| | - Michelle C. Chandley
- Biomedical Science Department, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
13
|
Wang Z, Li L, Li M, Lu Z, Qin L, Naumann RK, Wang H. Chemogenetic Modulation of Preoptic Gabre Neurons Decreases Body Temperature and Heart Rate. Int J Mol Sci 2024; 25:13061. [PMID: 39684772 DOI: 10.3390/ijms252313061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/18/2024] Open
Abstract
The preoptic area of the hypothalamus is critical for regulation of brain-body interaction, including circuits that control vital signs such as body temperature and heart rate. The preoptic area contains approximately 70 molecularly distinct cell types. The Gabre gene is expressed in a subset of preoptic area cell types. It encodes the GABA receptor ε-subunit, which is thought to confer resistance to anesthetics at the molecular level, but the function of Gabre cells in the brain remains largely unknown. We generated and have extensively characterized a Gabre-cre knock-in mouse line and used chemogenetic tools to interrogate the function of Gabre cells in the preoptic area. Comparison with macaque GABRE expression revealed the conserved character of Gabre cells in the preoptic area. In awake mice, we found that chemogenetic activation of Gabre neurons in the preoptic area reduced body temperature, whereas chemogenetic inhibition had no effect. Furthermore, chemogenetic inhibition of Gabre neurons in the preoptic area decreased the heart rate, whereas chemogenetic activation had no effect under isoflurane anesthesia. These findings suggest an important role of preoptic Gabre neurons in maintaining vital signs such as body temperature and heart rate during wakefulness and under anesthesia.
Collapse
Affiliation(s)
- Ziyue Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lanxiang Li
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miao Li
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Zhonghua Lu
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lihua Qin
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Robert Konrad Naumann
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Hong Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| |
Collapse
|
14
|
Lawton SB, Wagner VA, Nakagawa P, Segar JL, Sigmund CD, Morselli LL, Grobe JL. Angiotensin in the Arcuate: Mechanisms Integrating Cardiometabolic Control: The 2022 COH Mid-Career Award for Research Excellence. Hypertension 2024; 81:2209-2217. [PMID: 39315447 PMCID: PMC11483214 DOI: 10.1161/hypertensionaha.124.20524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The American Heart Association has identified obesity as a primary impediment to ongoing improvements in cardiovascular diseases, including hypertension. Although drugs, exercise, diets, and surgeries can each cause weight loss, few subjects maintain a reduced weight over the long term. Dysfunctional integrative control (ie, adaptation) of resting metabolic rate (RMR) appears to underlie this failed weight maintenance, yet the neurobiology of physiological and pathophysiological RMR control is poorly understood. Here, we review recent insights into the cellular and molecular control of RMR by Ang-II (angiotensin II) signaling within the arcuate nucleus of the hypothalamus. Within a unique subset of agouti-related peptide neurons, AT1R (Ang-II type 1 receptors) are implicated in the integrative control of RMR. Furthermore, a spontaneous G protein signal switch of AT1R within this neuron type appears to underlie the pathogenesis of RMR adaptation by qualitatively changing the cellular response to AT1R activation from a β-arrestin-1/Gαi (heterotrimeric G protein, α i subtype)-mediated inhibitory response to a Gαq (heterotrimeric G protein, α q subtype)-mediated stimulatory response. We conclude that therapeutic approaches to obesity are likely hampered by the plasticity of the signaling mechanisms that mediate the normal integrative control of energy balance. The same stimulus that would increase RMR in the normal physiological state may decrease RMR during obesity due to qualitative changes in second-messenger coupling. Understanding the mechanisms that regulate interactions between receptors such as AT1R and its various second messenger signaling cascades will provide novel insights into the pathogenesis of RMR adaptation and potentially point toward new therapeutic approaches for obesity and hypertension.
Collapse
Affiliation(s)
- Samuel B.R. Lawton
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Valerie A. Wagner
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jeffrey L. Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Lisa L. Morselli
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
15
|
El-Ansary MRM, El-Ansary AR, Said SM, Abdel-Hakeem MA. Regular cold shower exposure modulates humoral and cell-mediated immunity in healthy individuals. J Therm Biol 2024; 125:103971. [PMID: 39299098 DOI: 10.1016/j.jtherbio.2024.103971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Cold hydrotherapy is an ancient practice that has recently gained scientific interest for its potential health benefits. This study explored the effects of regular cold shower exposure on immune cell function. METHODS Sixty healthy Egyptian adults were randomized to take cold or hot showers daily for 90 days. Levels of immunoglobulins, cytokines, and interferon-gamma were measured in blood samples at baseline, 30, 60, and 90 days. RESULTS The cold shower group exhibited significant increases in immunoglobulin levels. Conversely, the hot shower group showed a significant decrease in IgM levels at 60 and 90 days compared to baseline, alongside nonsignificant decrease of IgG and IgA. the cold shower group demonstrated elevated levels of IL-2 and IL-4 at 90 days, indicating enhanced T-cell proliferation and humoral immunity, respectively. In contrast, the hot shower group did not exhibit significant changes in cytokine levels. There were no significant differences in IFN-γ and TNF-α levels between the groups. CONCLUSIONS Regular cold shower exposure appears to enhance humoral and cell-mediated immunity through the upregulation of antibodies, interleukin-2, and interleukin-4. Brief cold stressors may induce physiological adaptations that prime the immune response. This accessible, sustainable lifestyle modification could potentially serve as an alternative therapy to boost immunity. Further research on larger populations is warranted to better understand the physiological effects of cold temperatures on immunity.
Collapse
Affiliation(s)
- Mahmoud R M El-Ansary
- Department of Immunology and Medical Microbiology, Faculty of Medicine, Misr University for Science and Technology, Giza, Egypt
| | - Amira R El-Ansary
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Giza, Egypt
| | - Shereen M Said
- Department of Basic Science, Faculty of Physical Therapy, Misr University for Science and Technology, Giza, Egypt
| | - Mohamed A Abdel-Hakeem
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza, Egypt.
| |
Collapse
|
16
|
Mualem R, Morales-Quezada L, Farraj RH, Shance S, Bernshtein DH, Cohen S, Mualem L, Salem N, Yehuda RR, Zbedat Y, Waksman I, Biswas S. Econeurobiology and brain development in children: key factors affecting development, behavioral outcomes, and school interventions. Front Public Health 2024; 12:1376075. [PMID: 39391155 PMCID: PMC11465878 DOI: 10.3389/fpubh.2024.1376075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/29/2024] [Indexed: 10/12/2024] Open
Abstract
The Econeurobiology of the brain describes the environment in which an individual's brain develops. This paper explores the complex neural mechanisms that support and evaluate enrichment at various stages of development, providing an overview of how they contribute to plasticity and enhancement of both achievement and health. It explores the deep benefits of enrichment and contrasts them with the negative effects of trauma and stress on brain development. In addition, the paper strongly emphasizes the integration of Gardner's intelligence types into the school curriculum environment. It emphasizes the importance of linking various intelligence traits to educational strategies to ensure a holistic approach to cognitive development. In the field of Econeurobiology, this work explains the central role of the environment in shaping the development of the brain. It examines brain connections and plasticity and reveals the impact of certain environmental factors on brain development in early and mid-childhood. In particular, the six key factors highlighted are an environment of support, nutrition, physical activity, music, sleep, and cognitive strategies, highlighting their potential to improve cognitive abilities, memory, learning, self-regulation, and social and emotional development. This paper also investigates the social determinants of health and education in the context of Econeurobiology. It emphasizes the transformative power of education in society, especially in vulnerable communities facing global challenges in accessing quality education.
Collapse
Affiliation(s)
- Raed Mualem
- Department of Natural and Environmental Sciences, Faculty of Education, Oranim Academic College, Kiryat Tiv'on, Israel
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
- Ramat Zevulun High School, Ibtin, Israel
| | - Leon Morales-Quezada
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - Rania Hussein Farraj
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
| | - Shir Shance
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
| | | | - Sapir Cohen
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
| | - Loay Mualem
- Department of Computer Science, Haifa University, Haifa, Israel
| | - Niven Salem
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
| | - Rivka Riki Yehuda
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
| | | | - Igor Waksman
- Bar Ilan University Medical School, Tzfat, Israel
| | - Seema Biswas
- Global Health Research Laboratory, Department of Surgery B, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
17
|
Wang T, Teng B, Yao DR, Gao W, Oka Y. Organ-specific Sympathetic Innervation Defines Visceral Functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613934. [PMID: 39345605 PMCID: PMC11430017 DOI: 10.1101/2024.09.19.613934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The autonomic nervous system orchestrates the brain and body functions through the sympathetic and parasympathetic pathways. However, our understanding of the autonomic system, especially the sympathetic system, at the cellular and molecular levels is severely limited. Here, we show unique topological representations of individual visceral organs in the major abdominal sympathetic ganglion complex. Using multi-modal transcriptomic analyses, we identified distinct sympathetic populations that are both molecularly and spatially separable in the celiac-superior mesenteric ganglia (CG-SMG). Notably, individual CG-SMG populations exhibit selective and mutually exclusive axonal projections to visceral organs, targeting either the gastrointestinal (GI) tract or secretory areas including the pancreas and bile tract. This combinatorial innervation pattern suggests functional segregation between different CG-SMG populations. Indeed, our neural perturbation experiments demonstrated that one class of neurons selectively regulates GI food transit. Another class of neurons controls digestion and glucagon secretion independent of gut motility. These results reveal the molecularly diverse sympathetic system and suggest modular regulations of visceral organ functions through distinct sympathetic populations.
Collapse
|
18
|
Long P, Ma Q, Wang Z, Wang G, Jiang J, Gao L. Genetic patterning in hippocampus of rat undergoing impaired spatial memory induced by long-term heat stress. Heliyon 2024; 10:e37319. [PMID: 39296065 PMCID: PMC11408118 DOI: 10.1016/j.heliyon.2024.e37319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
The organism's normal physiological function is greatly impacted in a febrile environment, leading to the manifestation of pathological conditions including elevated body temperature, dehydration, gastric bleeding, and spermatogenic dysfunction. Numerous lines of evidence indicate that heat stress significantly impacts the brain's structure and function. Previous studies have demonstrated that both animals and humans experience cognitive impairment as a result of exposure to high temperatures. However, there is a lack of research on the effects of prolonged exposure to high-temperature environments on learning and memory function, as well as the underlying molecular regulatory mechanisms. In this study, we examined the impact of long-term heat stress exposure on spatial memory function in rats and conducted transcriptome sequencing analysis of rat hippocampal tissues to identify the crucial molecular targets affected by prolonged heat stress exposure. It was found that the long-term heat stress impaired rats' spatial memory function due to the pathological damages and apoptosis of hippocampal neurons at the CA3 region, which is accompanied with the decrease of growth hormone level in peripheral blood. RNA sequencing analysis revealed the signaling pathways related to positive regulation of external stimulation response and innate immune response were dramatically affected by heat stress. Among the verified differentially expressed genes, the knockdown of Arhgap36 in neuronal cell line HT22 significantly enhances the cell apoptosis, suggesting the impaired spatial memory induced by long-term heat stress may at least partially be mediated by the dysregulation of Arhgap36 in hippocampal neurons. The uncovered relationship between molecular changes in the hippocampus and behavioral alterations induced by long-term heat stress may offer valuable insights for the development of therapeutic targets and protective drugs to enhance memory function in heat-exposed individuals.
Collapse
Affiliation(s)
- Peihua Long
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Qunfei Ma
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Zhe Wang
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Guanqin Wang
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Jianan Jiang
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Lu Gao
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200120, PR China
| |
Collapse
|
19
|
Xu JH, He TH, Wang NP, Gao WM, Cheng YJ, Ji QF, Wu SH, Wei YL, Tang Y, Yang WZ, Zhang J. Thermoregulatory pathway underlying the pyrogenic effects of prostaglandin E 2 in the lateral parabrachial nucleus of male rats. Acta Pharmacol Sin 2024; 45:1832-1847. [PMID: 38702500 PMCID: PMC11336216 DOI: 10.1038/s41401-024-01289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
It has been shown that prostaglandin (PG) E2 synthesized in the lateral parabrachial nucleus (LPBN) is involved in lipopolysaccharide-induced fever. But the neural mechanisms of how intra-LPBN PGE2 induces fever remain unclear. In this study, we investigated whether the LPBN-preoptic area (POA) pathway, the thermoafferent pathway for feed-forward thermoregulatory responses, mediates fever induced by intra-LPBN PGE2 in male rats. The core temperature (Tcore) was monitored using a temperature radiotelemetry transponder implanted in rat abdomen. We showed that microinjection of PGE2 (0.28 nmol) into the LPBN significantly enhanced the density of c-Fos-positive neurons in the median preoptic area (MnPO). The chemical lesioning of MnPO with ibotenate or selective genetic lesioning or inhibition of the LPBN-MnPO pathway significantly attenuated fever induced by intra-LPBN injection of PGE2. We demonstrated that EP3 receptor was a pivotal receptor for PGE2-induced fever, since microinjection of EP3 receptor agonist sulprostone (0.2 nmol) or EP3 receptor antagonist L-798106 (2 nmol) into the LPBN mimicked or weakened the pyrogenic action of LPBN PGE2, respectively, but this was not the case for EP4 and EP1 receptors. Whole-cell recording from acute LPBN slices revealed that the majority of MnPO-projecting neurons originating from the external lateral (el) and dorsal (d) LPBN were excited and inhibited, respectively, by PGE2 perfusion, initiating heat-gain and heat-loss mechanisms. The amplitude but not the frequency of spontaneous and miniature glutamatergic excitatory postsynaptic currents (sEPSCs and mEPSCs) in MnPO-projecting LPBel neurons increased after perfusion with PGE2; whereas the frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) and the A-type potassium (IA) current density did not change. In MnPO-projecting LPBd neurons, neither sEPSCs nor sIPSCs responded to PGE2; however, the IA current density was significantly increased by PGE2 perfusion. These electrophysiological responses and the thermoeffector reactions to intra-LPBN PGE2 injection, including increased brown adipose tissue thermogenesis, shivering, and decreased heat dissipation, were all abolished by L-798106, and mimicked by sulprostone. These results suggest that the pyrogenic effects of intra-LPBN PGE2 are mediated by both the inhibition of the LPBd-POA pathway through the EP3 receptor-mediated activation of IA currents and the activation of the LPBel-POA pathway through the selective enhancement of glutamatergic synaptic transmission via EP3 receptors.
Collapse
Affiliation(s)
- Jian-Hui Xu
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Tian-Hui He
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Nan-Ping Wang
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Wen-Min Gao
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Yong-Jing Cheng
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Qiao-Feng Ji
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Si-Hao Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
| | - Yan-Lin Wei
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
| | - Yu Tang
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China
| | - Wen Z Yang
- School of Life Science and Technology, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Jie Zhang
- Key Laboratory of Thermoregulation and Inflammation of Sichuan Higher Education Institutes, Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
20
|
Liu H, Bean JC, Li Y, Yu M, Ginnard OZ, Conde KM, Wang M, Fang X, Liu H, Tu L, Yin N, Han J, Yang Y, Tong Q, Arenkiel BR, Wang C, He Y, Xu Y. Distinct basal forebrain-originated neural circuits promote homoeostatic feeding and suppress hedonic feeding in male mice. Nat Metab 2024; 6:1775-1790. [PMID: 39112722 PMCID: PMC11881791 DOI: 10.1038/s42255-024-01099-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/08/2024] [Indexed: 09/26/2024]
Abstract
Feeding behaviour is influenced by two primary factors: homoeostatic needs driven by hunger and hedonic desires for pleasure even in the absence of hunger. While efficient homoeostatic feeding is vital for survival, excessive hedonic feeding can lead to adverse consequences such as obesity and metabolic dysregulations. However, the neurobiological mechanisms that orchestrate homoeostatic versus hedonic food consumption remain largely unknown. Here we show that GABAergic proenkephalin (Penk) neurons in the diagonal band of Broca (DBB) of male mice respond to food presentation. We further demonstrate that a subset of DBBPenk neurons that project to the paraventricular nucleus of the hypothalamus are preferentially activated upon food presentation during fasting periods and transmit a positive valence to facilitate feeding. On the other hand, a separate subset of DBBPenk neurons that project to the lateral hypothalamus are preferentially activated when detecting a high-fat high-sugar (HFHS) diet and transmit a negative valence to inhibit food consumption. Notably, when given free choice of chow and HFHS diets, mice with the whole DBBPenk population ablated exhibit reduced consumption of chow but increased intake of the HFHS diet, resulting in accelerated development of obesity and metabolic disturbances. Together, we identify a molecularly defined neural population in male mice that is crucial for the maintenance of energy balance by facilitating homoeostatic feeding while suppressing hedonic overeating.
Collapse
Affiliation(s)
- Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan C Bean
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongxiang Li
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Olivia Z Ginnard
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kristine M Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Xing Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Na Yin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
Tabarean IV. Opposing actions of co-released GABA and neurotensin on the activity of preoptic neurons and on body temperature. eLife 2024; 13:RP98677. [PMID: 39207910 PMCID: PMC11361704 DOI: 10.7554/elife.98677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Neurotensin (Nts) is a neuropeptide acting as a neuromodulator in the brain. Pharmacological studies have identified Nts as a potent hypothermic agent. The medial preoptic area, a region that plays an important role in the control of thermoregulation, contains a high density of neurotensinergic neurons and Nts receptors. The conditions in which neurotensinergic neurons play a role in thermoregulation are not known. In this study, optogenetic stimulation of preoptic Nts neurons induced a small hyperthermia. In vitro, optogenetic stimulation of preoptic Nts neurons resulted in synaptic release of GABA and net inhibition of the preoptic pituitary adenylate cyclase-activating polypeptide (Adcyap1) neurons firing activity. GABA-A receptor antagonist or genetic deletion of Slc32a1 (VGAT) in Nts neurons unmasked also an excitatory effect that was blocked by a Nts receptor 1 antagonist. Stimulation of preoptic Nts neurons lacking Slc32a1 resulted in excitation of Adcyap1 neurons and hypothermia. Mice lacking Slc32a1 expression in Nts neurons presented changes in the fever response and in the responses to heat or cold exposure as well as an altered circadian rhythm of body temperature. Chemogenetic activation of all Nts neurons in the brain induced a 4-5°C hypothermia, which could be blocked by Nts receptor antagonists in the preoptic area. Chemogenetic activation of preoptic neurotensinergic projections resulted in robust excitation of preoptic Adcyap1 neurons. Taken together, our data demonstrate that endogenously released Nts can induce potent hypothermia and that excitation of preoptic Adcyap1 neurons is the cellular mechanism that triggers this response.
Collapse
|
22
|
Yang X, Qiu K, Jiang Y, Huang Y, Zhang Y, Liao Y. Metabolic Crosstalk between Liver and Brain: From Diseases to Mechanisms. Int J Mol Sci 2024; 25:7621. [PMID: 39062868 PMCID: PMC11277155 DOI: 10.3390/ijms25147621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple organs and tissues coordinate to respond to dietary and environmental challenges. It is interorgan crosstalk that contributes to systemic metabolic homeostasis. The liver and brain, as key metabolic organs, have their unique dialogue to transmit metabolic messages. The interconnected pathogenesis of liver and brain is implicated in numerous metabolic and neurodegenerative disorders. Recent insights have positioned the liver not only as a central metabolic hub but also as an endocrine organ, capable of secreting hepatokines that transmit metabolic signals throughout the body via the bloodstream. Metabolites from the liver or gut microbiota also facilitate a complex dialogue between liver and brain. In parallel to humoral factors, the neural pathways, particularly the hypothalamic nuclei and autonomic nervous system, are pivotal in modulating the bilateral metabolic interplay between the cerebral and hepatic compartments. The term "liver-brain axis" vividly portrays this interaction. At the end of this review, we summarize cutting-edge technical advancements that have enabled the observation and manipulation of these signals, including genetic engineering, molecular tracing, and delivery technologies. These innovations are paving the way for a deeper understanding of the liver-brain axis and its role in metabolic homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
23
|
Grajales-Reyes JG, Chen B, Meseguer D, Schneeberger M. Burning Question: How Does Our Brain Process Positive and Negative Cues Associated with Thermosensation? Physiology (Bethesda) 2024; 39:0. [PMID: 38536114 PMCID: PMC11368520 DOI: 10.1152/physiol.00034.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/22/2024] [Indexed: 05/16/2024] Open
Abstract
Whether it is the dramatic suffocating sensation from a heat wave in the summer or the positive reinforcement arising from a hot drink on a cold day; we can certainly agree that our thermal environment underlies our daily rhythms of sensation. Extensive research has focused on deciphering the central circuits responsible for conveying the impact of thermogenesis on mammalian behavior. Here, we revise the recent literature responsible for defining the behavioral correlates that arise from thermogenic fluctuations in mammals. We transition from the physiological significance of thermosensation to the circuitry responsible for the autonomic or behavioral responses associated with it. Subsequently, we delve into the positive and negative valence encoded by thermoregulatory processes. Importantly, we emphasize the crucial junctures where reward, pain, and thermoregulation intersect, unveiling a complex interplay within these neural circuits. Finally, we briefly outline fundamental questions that are pending to be addressed in the field. Fully deciphering the thermoregulatory circuitry in mammals will have far-reaching medical implications. For instance, it may lead to the identification of novel targets to overcome thermal pain or allow the maintenance of our core temperature in prolonged surgeries.
Collapse
Affiliation(s)
- Jose G Grajales-Reyes
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Bandy Chen
- Department of Cellular and Molecular Physiology, Laboratory of Neurovascular Control of Homeostasis, Yale School of Medicine, New Haven, Connecticut, United States
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, Connecticut, United States
| | - David Meseguer
- Department of Cellular and Molecular Physiology, Laboratory of Neurovascular Control of Homeostasis, Yale School of Medicine, New Haven, Connecticut, United States
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, Connecticut, United States
| | - Marc Schneeberger
- Department of Cellular and Molecular Physiology, Laboratory of Neurovascular Control of Homeostasis, Yale School of Medicine, New Haven, Connecticut, United States
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, Connecticut, United States
| |
Collapse
|
24
|
Ahmadlou M, Giannouli M, van Vierbergen JFM, van Leeuwen T, Bloem W, Houba JHW, Shirazi MY, Cazemier JL, Haak R, Dubey M, de Winter F, Heimel JA. Cell-type-specific hypothalamic pathways to brainstem drive context-dependent strategies in response to stressors. Curr Biol 2024; 34:2448-2459.e4. [PMID: 38754425 DOI: 10.1016/j.cub.2024.04.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/18/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Adaptive behavioral responses to stressors are critical for survival. However, which brain areas orchestrate switching the appropriate stress responses to distinct contexts is an open question. This study aimed to identify the cell-type-specific brain circuitry governing the selection of distinct behavioral strategies in response to stressors. Through novel mouse behavior paradigms, we observed distinct stressor-evoked behaviors in two psycho-spatially distinct contexts characterized by stressors inside or outside the safe zone. The identification of brain regions activated in both conditions revealed the involvement of the dorsomedial hypothalamus (DMH). Further investigation using optogenetics, chemogenetics, and photometry revealed that glutamatergic projections from the DMH to periaqueductal gray (PAG) mediated responses to inside stressors, while GABAergic projections, particularly from tachykinin1-expressing neurons, played a crucial role in coping with outside stressors. These findings elucidate the role of cell-type-specific circuitry from the DMH to the PAG in shaping behavioral strategies in response to stressors. These findings have the potential to advance our understanding of fundamental neurobiological processes and inform the development of novel approaches for managing context-dependent and anxiety-associated pathological conditions such as agoraphobia and claustrophobia.
Collapse
Affiliation(s)
- Mehran Ahmadlou
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, W1T4AJ London, UK.
| | - Maria Giannouli
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Jacqueline F M van Vierbergen
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Tom van Leeuwen
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Wouter Bloem
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Janou H W Houba
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Maryam Yasamin Shirazi
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - J Leonie Cazemier
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Robin Haak
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Mohit Dubey
- Department of Axonal Signaling, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Fred de Winter
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - J Alexander Heimel
- Circuits, Structure and Function Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Rogers JF, Vandendoren M, Prather JF, Landen JG, Bedford NL, Nelson AC. Neural cell-types and circuits linking thermoregulation and social behavior. Neurosci Biobehav Rev 2024; 161:105667. [PMID: 38599356 PMCID: PMC11163828 DOI: 10.1016/j.neubiorev.2024.105667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Understanding how social and affective behavioral states are controlled by neural circuits is a fundamental challenge in neurobiology. Despite increasing understanding of central circuits governing prosocial and agonistic interactions, how bodily autonomic processes regulate these behaviors is less resolved. Thermoregulation is vital for maintaining homeostasis, but also associated with cognitive, physical, affective, and behavioral states. Here, we posit that adjusting body temperature may be integral to the appropriate expression of social behavior and argue that understanding neural links between behavior and thermoregulation is timely. First, changes in behavioral states-including social interaction-often accompany changes in body temperature. Second, recent work has uncovered neural populations controlling both thermoregulatory and social behavioral pathways. We identify additional neural populations that, in separate studies, control social behavior and thermoregulation, and highlight their relevance to human and animal studies. Third, dysregulation of body temperature is linked to human neuropsychiatric disorders. Although body temperature is a "hidden state" in many neurobiological studies, it likely plays an underappreciated role in regulating social and affective states.
Collapse
Affiliation(s)
- Joseph F Rogers
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Morgane Vandendoren
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Jonathan F Prather
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Jason G Landen
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Nicole L Bedford
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Adam C Nelson
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA.
| |
Collapse
|
26
|
Luo X, Li DD, Li ZC, Li ZX, Zou DH, Huang F, Wang G, Wang R, Cao YF, Sun WY, Kurihara H, Liang L, Li YF, Jin W, Wu YP, He RR. Mitigating phospholipid peroxidation of macrophages in stress-induced tumor microenvironment by natural ALOX15/PEBP1 complex inhibitors. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155475. [PMID: 38492368 DOI: 10.1016/j.phymed.2024.155475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND The intricate interactions between chronic psychological stress and susceptibility to breast cancer have been recognized, yet the underlying mechanisms remain unexplored. Danzhi Xiaoyao Powder (DZXY), a traditional Chinese medicine (TCM) formula, has found clinical utility in the treatment of breast cancer. Macrophages, as the predominant immune cell population within the tumor microenvironment (TME), play a pivotal role in orchestrating tumor immunosurveillance. Emerging evidence suggests that lipid oxidation accumulation in TME macrophages, plays a critical role in breast cancer development and progression. However, a comprehensive understanding of the pharmacological mechanisms and active components of DZXY related to its clinical application in the treatment of stress-aggravated breast cancer remains elusive. PURPOSE This study sought to explore the plausible regulatory mechanisms and identify the key active components of DZXY contributing to its therapeutic efficacy in the context of breast cancer. METHODS Initially, we conducted an investigation into the relationship between the phagocytic capacity of macrophages damaged by psychological stress and phospholipid peroxidation using flow cytometry and LC-MS/MS-based phospholipomics. Subsequently, we evaluated the therapeutic efficacy of DZXY based on the results of the tumor size, tumor weight, the phospholipid peroxidation pathway and phagocytosis of macrophage. Additionally, the target-mediated characterization strategy based on binding of arachidonate 15-lipoxygenase (ALOX15) to phosphatidylethanolamine-binding protein-1 (PEBP1), including molecular docking analysis, microscale thermophoresis (MST) assay, co-immunoprecipitation analysis and activity verification, has been further implemented to reveal the key bio-active components in DZXY. Finally, we evaluated the therapeutic efficacy of isochlorogenic acid C (ICAC) based on the results of tumor size, tumor weight, the phospholipid peroxidation pathway, and macrophage phagocytosis in vivo. RESULTS The present study demonstrated that phospholipid peroxides, as determined by LC-MS/MS-based phospholipidomics, triggered in macrophages, which in turn compromised their capacity to eliminate tumor cells through phagocytosis. Furthermore, we elucidate the mechanism behind stress-induced PEBP1 to form a complex with ALOX15, thereby mediating membrane phospholipid peroxidation in macrophages. DZXY, demonstrates potent anti-breast cancer therapeutic effects by disrupting the ALOX15/PEBP1 interaction and inhibiting phospholipid peroxidation, ultimately enhancing macrophages' phagocytic capability towards tumor cells. Notably, ICAC emerged as a promising active component in DZXY, which can inhibit the ALOX15/PEBP1 interaction, thereby mitigating phospholipid peroxidation in macrophages. CONCLUSION Collectively, our findings elucidate stress increases the susceptibility of breast cancer by driving lipid peroxidation of macrophages and suggest the ALOX15/PEBP1 complex as a promising intervention target for DZXY.
Collapse
Affiliation(s)
- Xiang Luo
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Dong-Dong Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zi-Chun Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zi-Xuan Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - De-Hua Zou
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Feng Huang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Rong Wang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yun-Feng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, NHC Key Laboratory of Reproduction Regulation, Shang Hai 200032, China
| | - Wan-Yang Sun
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Hiroshi Kurihara
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Lei Liang
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Yi-Fang Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Wen Jin
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China.
| | - Yan-Ping Wu
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China.
| | - Rong-Rong He
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/Integrated Chinese and Western Medicine Postdoctoral Research Station/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China.
| |
Collapse
|
27
|
Oya M, Miyasaka Y, Nakamura Y, Tanaka M, Suganami T, Mashimo T, Nakamura K. Age-related ciliopathy: Obesogenic shortening of melanocortin-4 receptor-bearing neuronal primary cilia. Cell Metab 2024; 36:1044-1058.e10. [PMID: 38452767 DOI: 10.1016/j.cmet.2024.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 01/16/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Obesity is often associated with aging. However, the mechanism of age-related obesity is unknown. The melanocortin-4 receptor (MC4R) mediates leptin-melanocortin anti-obesity signaling in the hypothalamus. Here, we discovered that MC4R-bearing primary cilia of hypothalamic neurons progressively shorten with age in rats, correlating with age-dependent metabolic decline and increased adiposity. This "age-related ciliopathy" is promoted by overnutrition-induced upregulation of leptin-melanocortin signaling and inhibited or reversed by dietary restriction or the knockdown of ciliogenesis-associated kinase 1 (CILK1). Forced shortening of MC4R-bearing cilia in hypothalamic neurons by genetic approaches impaired neuronal sensitivity to melanocortin and resulted in decreased brown fat thermogenesis and energy expenditure and increased appetite, finally developing obesity and leptin resistance. Therefore, despite its acute anti-obesity effect, chronic leptin-melanocortin signaling increases susceptibility to obesity by promoting the age-related shortening of MC4R-bearing cilia. This study provides a crucial mechanism for age-related obesity, which increases the risk of metabolic syndrome.
Collapse
Affiliation(s)
- Manami Oya
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yoshiki Miyasaka
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yoshiko Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan; Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya 464-8601, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan; Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya 464-8601, Japan; Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Nagoya 464-8601, Japan
| | - Tomoji Mashimo
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Division of Genome Engineering, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
28
|
Botterill JJ, Khlaifia A, Appings R, Wilkin J, Violi F, Premachandran H, Cruz-Sanchez A, Canella AE, Patel A, Zaidi SD, Arruda-Carvalho M. Dorsal peduncular cortex activity modulates affective behavior and fear extinction in mice. Neuropsychopharmacology 2024; 49:993-1006. [PMID: 38233571 PMCID: PMC11039686 DOI: 10.1038/s41386-024-01795-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
The medial prefrontal cortex (mPFC) is critical to cognitive and emotional function and underlies many neuropsychiatric disorders, including mood, fear and anxiety disorders. In rodents, disruption of mPFC activity affects anxiety- and depression-like behavior, with specialized contributions from its subdivisions. The rodent mPFC is divided into the dorsomedial prefrontal cortex (dmPFC), spanning the anterior cingulate cortex (ACC) and dorsal prelimbic cortex (PL), and the ventromedial prefrontal cortex (vmPFC), which includes the ventral PL, infralimbic cortex (IL), and in some studies the dorsal peduncular cortex (DP) and dorsal tenia tecta (DTT). The DP/DTT have recently been implicated in the regulation of stress-induced sympathetic responses via projections to the hypothalamus. While many studies implicate the PL and IL in anxiety-, depression-like and fear behavior, the contribution of the DP/DTT to affective and emotional behavior remains unknown. Here, we used chemogenetics and optogenetics to bidirectionally modulate DP/DTT activity and examine its effects on affective behaviors, fear and stress responses in C57BL/6J mice. Acute chemogenetic activation of DP/DTT significantly increased anxiety-like behavior in the open field and elevated plus maze tests, as well as passive coping in the tail suspension test. DP/DTT activation also led to an increase in serum corticosterone levels and facilitated auditory fear extinction learning and retrieval. Activation of DP/DTT projections to the dorsomedial hypothalamus (DMH) acutely decreased freezing at baseline and during extinction learning, but did not alter affective behavior. These findings point to the DP/DTT as a new regulator of affective behavior and fear extinction in mice.
Collapse
Affiliation(s)
- Justin J Botterill
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Abdessattar Khlaifia
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Ryan Appings
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Jennifer Wilkin
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Francesca Violi
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Hanista Premachandran
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Arely Cruz-Sanchez
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada
| | - Anna Elisabete Canella
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Ashutosh Patel
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - S Danyal Zaidi
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada.
| |
Collapse
|
29
|
Campos-Cardoso R, Desa ZR, Fitzgerald BL, Moore AG, Duhon JL, Landar VA, Clem RL, Cummings KA. The mouse dorsal peduncular cortex encodes fear memory. Cell Rep 2024; 43:114097. [PMID: 38613783 PMCID: PMC11135038 DOI: 10.1016/j.celrep.2024.114097] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/26/2024] [Accepted: 03/27/2024] [Indexed: 04/15/2024] Open
Abstract
The rodent medial prefrontal cortex (mPFC) is functionally organized across the dorsoventral axis, where dorsal and ventral subregions promote and suppress fear, respectively. As the ventral-most subregion, the dorsal peduncular cortex (DP) is hypothesized to function in fear suppression. However, this role has not been explicitly tested. Here, we demonstrate that the DP paradoxically functions as a fear-encoding brain region and plays a minimal role in fear suppression. By using multimodal analyses, we demonstrate that DP neurons exhibit fear-learning-related plasticity and acquire cue-associated activity across learning and memory retrieval and that DP neurons activated by fear memory acquisition are preferentially reactivated upon fear memory retrieval. Further, optogenetic activation and silencing of DP fear-related neural ensembles drive the promotion and suppression of freezing, respectively. Overall, our results suggest that the DP plays a role in fear memory encoding. Moreover, our findings redefine our understanding of the functional organization of the rodent mPFC.
Collapse
Affiliation(s)
- Rodrigo Campos-Cardoso
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Zephyr R Desa
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Brianna L Fitzgerald
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Alana G Moore
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jace L Duhon
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Victoria A Landar
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Roger L Clem
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kirstie A Cummings
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
30
|
Tabarean IV. Opposing actions of co-released GABA and neurotensin on the activity of preoptic neurons and on body temperature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589556. [PMID: 38659782 PMCID: PMC11042348 DOI: 10.1101/2024.04.15.589556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Neurotensin (Nts) is a neuropeptide acting as a neuromodulator in the brain. Pharmacological studies have identified Nts as a potent hypothermic agent. The medial preoptic area, a region that plays an important role in the control of thermoregulation, contains a high density of neurotensinergic neurons and Nts receptors. The conditions in which neurotensinergic neurons play a role in thermoregulation are not known. In this study optogenetic stimulation of preoptic Nts neurons induced a small hyperthermia. In vitro, optogenetic stimulation of preoptic Nts neurons resulted in synaptic release of GABA and net inhibition of the preoptic pituitary adenylate cyclase-activating polypeptide (PACAP) neurons firing activity. GABA-A receptor antagonist or genetic deletion of VGAT in Nts neurons unmasked also an excitatory effect that was blocked by a Nts receptor 1 antagonist. Stimulation of preoptic Nts neurons lacking VGAT resulted in excitation of PACAP neurons and hypothermia. Mice lacking VGAT expression in Nts neurons presented changes in the fever response and in the responses to heat or cold exposure as well as an altered circadian rhythm of body temperature. Chemogenetic activation of all Nts neurons in the brain induced a 4-5 °C hypothermia, which could be blocked by Nts receptor antagonists in the preoptic area. Chemogenetic activation of preoptic neurotensinergic projections resulted in robust excitation of preoptic PACAP neurons. Taken together our data demonstrate that endogenously released Nts can induce potent hypothermia and that excitation of preoptic PACAP neurons is the cellular mechanism that triggers this response.
Collapse
|
31
|
Tsuno Y, Mieda M. Circadian rhythm mechanism in the suprachiasmatic nucleus and its relation to the olfactory system. Front Neural Circuits 2024; 18:1385908. [PMID: 38590628 PMCID: PMC11000122 DOI: 10.3389/fncir.2024.1385908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Animals need sleep, and the suprachiasmatic nucleus, the center of the circadian rhythm, plays an important role in determining the timing of sleep. The main input to the suprachiasmatic nucleus is the retinohypothalamic tract, with additional inputs from the intergeniculate leaflet pathway, the serotonergic afferent from the raphe, and other hypothalamic regions. Within the suprachiasmatic nucleus, two of the major subtypes are vasoactive intestinal polypeptide (VIP)-positive neurons and arginine-vasopressin (AVP)-positive neurons. VIP neurons are important for light entrainment and synchronization of suprachiasmatic nucleus neurons, whereas AVP neurons are important for circadian period determination. Output targets of the suprachiasmatic nucleus include the hypothalamus (subparaventricular zone, paraventricular hypothalamic nucleus, preoptic area, and medial hypothalamus), the thalamus (paraventricular thalamic nuclei), and lateral septum. The suprachiasmatic nucleus also sends information through several brain regions to the pineal gland. The olfactory bulb is thought to be able to generate a circadian rhythm without the suprachiasmatic nucleus. Some reports indicate that circadian rhythms of the olfactory bulb and olfactory cortex exist in the absence of the suprachiasmatic nucleus, but another report claims the influence of the suprachiasmatic nucleus. The regulation of circadian rhythms by sensory inputs other than light stimuli, including olfaction, has not been well studied and further progress is expected.
Collapse
Affiliation(s)
- Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | | |
Collapse
|
32
|
Gu LH, Wu RR, Zheng XL, Fu A, Xing ZY, Chen YY, He ZC, Lu LZ, Qi YT, Chen AH, Zhang YP, Xu TS, Peng MS, Ma C. Genomic insights into local adaptation and phenotypic diversity of Wenchang chickens. Poult Sci 2024; 103:103376. [PMID: 38228059 PMCID: PMC10823079 DOI: 10.1016/j.psj.2023.103376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024] Open
Abstract
Wenchang chicken, a prized local breed in Hainan Province of China renowned for its exceptional adaptability to tropical environments and good meat quality, is deeply favored by the public. However, an insufficient understanding of its population architecture and the unclear genetic basis that governs its typical attributes have posed challenges in the protection and breeding of this precious breed. To address these gaps, we conducted whole-genome resequencing on 200 Wenchang chicken samples derived from 10 distinct strains, and we gathered data on an array of 21 phenotype traits. Population genomics analysis unveiled distinctive population structures in Wenchang chickens, primarily attributed to strong artificial selection for different feather colors. Selection sweep analysis identified a group of candidate genes, including PCDH9, DPF3, CDIN1, and SUGCT, closely linked to adaptations that enhance resilience in tropical island habitats. Genome-wide association studies (GWAS) highlighted potential candidate genes associated with diverse feather color traits, encompassing TYR, RAB38, TRPM1, GABARAPL2, CDH1, ZMIZ1, LYST, MC1R, and SASH1. Through the comprehensive analysis of high-quality genomic and phenotypic data across diverse Wenchang chicken resource groups, this study unveils the intricate genetic backgrounds and population structures of Wenchang chickens. Additionally, it identifies multiple candidate genes linked to environmental adaptation, feather color variations, and production traits. These insights not only provide genetic reference for the purification and breeding of Wenchang chickens but also broaden our understanding of the genetic basis of phenotypic diversity in chickens.
Collapse
Affiliation(s)
- Li-Hong Gu
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571199, China
| | - Ran-Ran Wu
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin-Li Zheng
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571199, China
| | - An Fu
- Wenchang City Wenchang Chicken Research Institute, Wenchang 571300, China
| | - Zeng-Yang Xing
- Wenchang Long-quan Wenchang Chicken Industrial Co., Ltd., Wenchang 571346, China
| | - Yi-Yong Chen
- Hainan Chuang Wen Wenchang Chicken Industry Co., Ltd., Wenchang 571321, China
| | - Zhong-Chun He
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571199, China
| | - Li-Zhi Lu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yan-Tao Qi
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571199, China
| | - An-Hong Chen
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571199, China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tie-Shan Xu
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Min-Sheng Peng
- Wenchang City Wenchang Chicken Research Institute, Wenchang 571300, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cheng Ma
- Wenchang City Wenchang Chicken Research Institute, Wenchang 571300, China.
| |
Collapse
|
33
|
Palieri V, Paoli E, Wu YK, Haesemeyer M, Grunwald Kadow IC, Portugues R. The preoptic area and dorsal habenula jointly support homeostatic navigation in larval zebrafish. Curr Biol 2024; 34:489-504.e7. [PMID: 38211586 PMCID: PMC10849091 DOI: 10.1016/j.cub.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Animals must maintain physiological processes within an optimal temperature range despite changes in their environment. Through behavioral assays, whole-brain functional imaging, and neural ablations, we show that larval zebrafish, an ectothermic vertebrate, achieves thermoregulation through homeostatic navigation-non-directional and directional movements toward the temperature closest to its physiological setpoint. A brain-wide circuit encompassing several brain regions enables this behavior. We identified the preoptic area of the hypothalamus (PoA) as a key brain structure in triggering non-directional reorientation when thermal conditions are worsening. This result shows an evolutionary conserved role of the PoA as principal thermoregulator of the brain also in ectotherms. We further show that the habenula (Hb)-interpeduncular nucleus (IPN) circuit retains a short-term memory of the sensory history to support the generation of coherent directed movements even in the absence of continuous sensory cues. We finally provide evidence that this circuit may not be exclusive for temperature but may convey a more abstract representation of relative valence of physiologically meaningful stimuli regardless of their specific identity to enable homeostatic navigation.
Collapse
Affiliation(s)
- Virginia Palieri
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany; School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Emanuele Paoli
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - You Kure Wu
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Martin Haesemeyer
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Ilona C Grunwald Kadow
- School of Life Sciences, Technical University of Munich, Freising, Germany; Institute of Physiology II, University of Bonn, Medical Faculty (UKB), Nussallee 11, 53115 Bonn, Germany.
| | - Ruben Portugues
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany.
| |
Collapse
|
34
|
Tsuneoka Y, Funato H. Whole Brain Mapping of Orexin Receptor mRNA Expression Visualized by Branched In Situ Hybridization Chain Reaction. eNeuro 2024; 11:ENEURO.0474-23.2024. [PMID: 38199807 PMCID: PMC10883752 DOI: 10.1523/eneuro.0474-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Orexins, which are produced within neurons of the lateral hypothalamic area, play a pivotal role in the regulation of various behaviors, including sleep/wakefulness, reward behavior, and energy metabolism, via orexin receptor type 1 (OX1R) and type 2 (OX2R). Despite the advanced understanding of orexinergic regulation of behavior at the circuit level, the precise distribution of orexin receptors in the brain remains unknown. Here, we develop a new branched in situ hybridization chain reaction (bHCR) technique to visualize multiple target mRNAs in a semiquantitative manner, combined with immunohistochemistry, which provided comprehensive distribution of orexin receptor mRNA and neuron subtypes expressing orexin receptors in mouse brains. Only a limited number of cells expressing both Ox1r and Ox2r were observed in specific brain regions, such as the dorsal raphe nucleus and ventromedial hypothalamic nucleus. In many brain regions, Ox1r-expressing cells and Ox2r-expressing cells belong to different cell types, such as glutamatergic and GABAergic neurons. Moreover, our findings demonstrated considerable heterogeneity in Ox1r- or Ox2r-expressing populations of serotonergic, dopaminergic, noradrenergic, cholinergic, and histaminergic neurons. The majority of orexin neurons did not express orexin receptors. This study provides valuable insights into the mechanism underlying the physiological and behavioral regulation mediated by the orexin system, as well as the development of therapeutic agents targeting orexin receptors.
Collapse
Affiliation(s)
- Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 145-854, Japan
| | - Hiromasa Funato
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 145-854, Japan
- International Institutes for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
35
|
Cardoso RC, Desa ZR, Fitzgerald BL, Moore A, Duhon J, Landar VA, Clem RL, Cummings KA. The mouse dorsal peduncular cortex encodes fear memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550408. [PMID: 37546717 PMCID: PMC10402043 DOI: 10.1101/2023.07.24.550408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The rodent medial prefrontal cortex (mPFC) is a locus for both the promotion and suppression (e.g. extinction) of fear and is composed of four anatomically distinct subregions, including anterior cingulate 1 (Cg1), prelimbic (PL), infralimbic (IL), and the dorsal peduncular (DP) cortex. A vast majority of studies have focused on Cg1, PL, and IL. The Cg1 and PL have been implicated in the promotion of fear, while the IL has been linked to a role in the suppression, or extinction, of fear. Due to its anatomical location ventral to IL, the DP has been hypothesized to function as a fear-suppressing brain region however, no studies have explicitly tested its role in this function or in the regulation of memory generally. Moreover, some studies have pointed towards a dichotomous role for ventral mPFC in the dual suppression and promotion of fear, but the mechanisms underlying these opposing observations remains unclear. Here, we provide evidence that the DP paradoxically functions as a cued fear-encoding brain region and plays little to no role in fear memory extinction. By using a combination of cFos immunohistochemistry, whole-cell brain slice electrophysiology, fiber photometry, and activity-dependent neural tagging, we demonstrate that DP neurons exhibit learning-related plasticity, acquire cue-associated activity across learning and memory retrieval, and that DP neurons activated by learning are preferentially reactivated upon fear memory retrieval. Further, optogenetic activation and silencing of fear learning-related DP neural ensembles drives the promotion and suppression of freezing, respectively. Overall, these data suggest that the DP plays an unexpected role in fear memory encoding. More broadly, our results reveal new principles of organization across the dorsoventral axis of the mPFC.
Collapse
|
36
|
Pang P, Zhou X, Hu Y, Zhang Y, He B, Xu G. Time-series analysis of meteorological factors and emergency department visits due to dog/cat bites in Jinshan area, China. PeerJ 2024; 12:e16758. [PMID: 38250715 PMCID: PMC10800098 DOI: 10.7717/peerj.16758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Background Meteorological factors play an important role in human health. Clarifying the occurrence of dog and cat bites (DCBs) under different meteorological conditions can provide key insights into the prevention of DCBs. Therefore, the objective of the study was to explore the relationship between meteorological factors and DCBs and to provide caution to avoid the incidents that may occur by DCBs. Methods In this study, data on meteorological factors and cases of DCBs were retrospectively collected at the Shanghai Climate Center and Jinshan Hospital of Fudan University, respectively, in 2016-2020. The distributed lag non-linear and time series model (DLNM) were used to examine the effect of meteorological elements on daily hospital visits due to DCBs. Results A total of 26,857 DCBs were collected ranging from 1 to 39 cases per day. The relationship between ambient temperature and DCBs was J-shaped. DCBs were positively correlated with daily mean temperature (rs = 0.588, P < 0.01). The relative risk (RR) of DCBs was associated with high temperature (RR = 1.450; 95% CI [1.220-1.722]). Female was more susceptible to high temperature than male. High temperature increased the risk of DCBs. Conclusions The extremely high temperature increased the risk of injuries caused by DCBs, particularly for females. These data may help to develop public health strategies for potentially avoiding the occurrence of DCBs.
Collapse
Affiliation(s)
- Pei Pang
- Department of Medical Affairs, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyong Zhou
- Department of Medical Affairs, Jinshan Hospital, Fudan University, Shanghai, China
- Emergency Department, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yabin Hu
- Key Lab of Health Technology Assessment, National Health Commission of the People’s Republic of China, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Yin Zhang
- Shanghai Meteorological Service Center, Shanghai, China
| | - Baoshi He
- Key Laboratory of Watershed Geographic Sciences, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing, China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Nakamura K. Central Mechanisms of Thermoregulation and Fever in Mammals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:141-159. [PMID: 39289279 DOI: 10.1007/978-981-97-4584-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Thermoregulation is a fundamental homeostatic function in mammals mediated by the central nervous system. The framework of the central circuitry for thermoregulation lies in the hypothalamus and brainstem. The preoptic area (POA) of the hypothalamus integrates cutaneous and central thermosensory information into efferent control signals that regulate excitatory descending pathways through the dorsomedial hypothalamus (DMH) and rostral medullary raphe region (rMR). The cutaneous thermosensory feedforward signals are delivered to the POA by afferent pathways through the lateral parabrachial nucleus, while the central monitoring of body core temperature is primarily mediated by warm-sensitive neurons in the POA for negative feedback regulation. Prostaglandin E2, a pyrogenic mediator produced in response to infection, acts on the POA to trigger fever. Recent studies have revealed that this circuitry also functions for physiological responses to psychological stress and starvation. Master psychological stress signaling from the medial prefrontal cortex to the DMH has been discovered to drive a variety of physiological responses for stress coping, including hyperthermia. During starvation, hunger signaling from the hypothalamus was found to activate medullary reticular neurons, which then suppress thermogenic sympathetic outflows from the rMR for energy saving. This thermoregulatory circuit represents a fundamental mechanism of the central regulation for homeostasis.
Collapse
Affiliation(s)
- Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
38
|
Rony MKK, Alamgir HM. High temperatures on mental health: Recognizing the association and the need for proactive strategies-A perspective. Health Sci Rep 2023; 6:e1729. [PMID: 38059052 PMCID: PMC10696165 DOI: 10.1002/hsr2.1729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/28/2023] [Accepted: 11/05/2023] [Indexed: 12/08/2023] Open
Abstract
Background and Aims The influence of temperature on various aspects of daily life is often underestimated, and its effects on mental health are not widely recognized. Understanding and addressing the relationship between temperature and mental well-being is crucial in the context of climate change and rising global temperatures. This perspective aimed to investigate the effects of high temperatures on mental health and identify proactive strategies to mitigate these effects. Methods This perspective adopted a twofold approach, including a comprehensive literature review and socioecological framework. The literature review involved extensive searches across Google Scholar, PubMed, and Scopus to identify relevant, peer-reviewed articles, and reports from diverse disciplines. Results The perspective emphasized the significance of recognizing heat stress and its consequences on mental well-being. Chronic heat stress can lead to increased stress, anxiety, and cognitive impairment. Vulnerable populations include, the very young, older adults, and individuals with pre-existing mental health conditions. Socioeconomic factors can further exacerbate vulnerability, highlighting the need for tailored strategies to manage mental health challenges during high temperatures. Additionally, the article identified and discussed proactive coping strategies to minimize both the psychological and physical impacts of heat stress. Mindfulness, stress management techniques, and therapy are suggested as effective means for individuals to manage psychological distress. Conclusion Implementing preventive measures are essential steps in promoting mental wellness in high temperatures. Proactive strategies by addressing the physiological and psychological effects of heat and considering the specific needs of vulnerable populations can help individuals and communities navigate the challenges posed by rising temperatures and promote resilience and preserve their mental well-being.
Collapse
Affiliation(s)
- Moustaq Karim Khan Rony
- Department of Public HealthBangladesh Open UniversityGazipurBangladesh
- Department of Institute of Social Welfare and ResearchUniversity of DhakaDhakaBangladesh
| | - Hasnat M. Alamgir
- Department of Career & Professional Development Services (CPDS)Southeast UniversityDhakaBangladesh
| |
Collapse
|
39
|
Oka T. Functional hyperthermia and comorbid psychiatric disorders. Biopsychosoc Med 2023; 17:39. [PMID: 37957731 PMCID: PMC10644480 DOI: 10.1186/s13030-023-00295-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Affiliation(s)
- Takakazu Oka
- Department of Psychosomatic Medicine, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita, 286-8520, Chiba, Japan.
| |
Collapse
|
40
|
Lee S, Tochinai R, Yasuoka A, Nagai T, Saito Y, Kuwahara M, Abe K, Asakura T. Mastication stimuli regulate the heartbeat rate through rhythmic regulation by the hypothalamic-autonomic system; molecular and telemetric studies in weaning-stage rats. Front Neurosci 2023; 17:1260655. [PMID: 37781249 PMCID: PMC10536135 DOI: 10.3389/fnins.2023.1260655] [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: 07/19/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Mastication stimuli have been demonstrated to affect memory function and autonomic nerve activity; however, this process has not been well studied during weaning compared to old age. Previously, we conducted molecular analyses of the thalamus and hippocampus to elucidate the mechanisms underlying this memory-enhancing effect in weaning-stage rats. In this study, we aimed to evaluate the effect of masticatory stimuli on the regulation of heartbeat rate (HR) through the hypothalamic-autonomic system. Three-week-old male rats were administered a powdered diet (P group) or chow-diet (C group) for 10 days. Thereafter, transcriptome analysis was performed. Vasopressin, cocaine-amphetamine-regulated transcript prepropeptide, corticotropin-releasing hormone, and thyrotropin-releasing hormone, which are involved in sympathetic activation of heart rate, were downregulated in the C group. Electrocardiograms were recorded continuously for 12 days under the same condition. Interestingly, rats in the C group had a significantly lower HR than those in the P group on day 11. We checked several parameters representing the autonomic regulation of HR. The C group had higher values for the high-frequency band integration of the HR power spectrum (parasympathetic marker) and root mean square successive difference of R-wave intervals (parasympathetic marker) relative to the P group. Such findings provide a molecular and physiological basis for understanding the regulation of cardiovascular function in response to masticatory stimuli in the autonomic nervous system.
Collapse
Affiliation(s)
- Seonmi Lee
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryota Tochinai
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Akihito Yasuoka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Human Nutrition, Seitoku University, Chiba, Japan
| | - Toshitada Nagai
- Department of Applied Biological Science, Faculty of Agriculture, Takasaki University of Health and Welfare, Gunma, Japan
| | - Yoshikazu Saito
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Toyo Institute of Food Technology, Hyogo, Japan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Keiko Abe
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Project on Health and Anti-Aging, Kanagawa Academy of Science and Technology, Life Science and Environment Research Center (LiSE), Kawasaki, Japan
| | - Tomiko Asakura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Grobe CC, Reho JJ, Brown-Williams D, Ziegler AA, Mathieu NM, Lawton SB, Fekete EM, Brozoski DT, Wackman KK, Burnett CM, Nakagawa P, Sigmund CD, Segar JL, Grobe JL. Cardiometabolic Effects of DOCA-Salt in Mice Depend on Ambient Temperature. Hypertension 2023; 80:1871-1880. [PMID: 37470185 PMCID: PMC10528934 DOI: 10.1161/hypertensionaha.122.20415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 07/10/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Mice prefer warmer environments than humans. For this reason, behavioral and physiological thermoregulatory responses are engaged by mice in response to a standard room temperature of 22 to 24 °C. Autonomic mechanisms mediating thermoregulatory responses overlap with mechanisms activated in hypertension, and, therefore, we hypothesized that housing at thermoneutral temperatures (TNs; 30 °C) would modify the cardiometabolic effects of deoxycorticosterone acetate (DOCA)-salt in mice. METHODS The effects of DOCA-salt treatment upon ingestive behaviors, energy expenditure, blood pressure, heart rate (HR), and core temperature were assessed in C57BL/6J mice housed at room temperature or TN. RESULTS Housing at TN reduced food intake, energy expenditure, blood pressure, and HR and attenuated HR responses to acute autonomic blockade by chlorisondamine. At room temperature, DOCA-salt caused expected increases in fluid intake, sodium retention in osmotically inactive pools, blood pressure, core temperature, and also caused expected decreases in fat-free mass, total body water, and HR. At TN, the effects of DOCA-salt upon fluid intake, fat gains, hydration, and core temperature were exaggerated, but effects on energy expenditure and HR were blunted. Effects of DOCA-salt upon blood pressure were similar for 3 weeks and exaggerated by TN housing in the fourth week. CONCLUSIONS Ambient temperature robustly influences behavioral and physiological functions in mice, including metabolic and cardiovascular phenotype development in response to DOCA-salt treatment. Studying cardiometabolic responses of mice at optimal ambient temperatures promises to improve the translational relevance of rodent models.
Collapse
Affiliation(s)
- Connie C. Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John J. Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
| | | | - Alisha A. Ziegler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Natalia M. Mathieu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Samuel B.R. Lawton
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Eva M. Fekete
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Daniel T. Brozoski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Kelsey K. Wackman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Colin M.L. Burnett
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jeffrey L. Segar
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
42
|
Zhu Y, He C, Bell M, Zhang Y, Fatmi Z, Zhang Y, Zaid M, Bachwenkizi J, Liu C, Zhou L, Chen R, Kan H. Association of Ambient Temperature With the Prevalence of Intimate Partner Violence Among Partnered Women in Low- and Middle-Income South Asian Countries. JAMA Psychiatry 2023; 80:952-961. [PMID: 37379013 PMCID: PMC10308303 DOI: 10.1001/jamapsychiatry.2023.1958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/24/2023] [Indexed: 06/29/2023]
Abstract
Importance Intimate partner violence (IPV), including physical, sexual, and emotional violence, constitutes a critical public health problem, particularly in low- and middle-income countries. While climate change could escalate violent events, data quantifying its possible association with IPV are scant. Objective To evaluate the association of ambient temperature with the prevalence of IPV among partnered women in low- and middle-income countries in South Asia, and to estimate the association of future climate warming with IPV. Design, Setting, and Participants This cross-sectional study used data from the Demographic and Health Survey and included 194 871 ever-partnered women aged 15 to 49 years from 3 South Asian countries (India, Nepal, and Pakistan). The study applied the mixed-effect multivariable logistic regression model to investigate the association of ambient temperature with IPV prevalence. The study further modeled the change in IPV prevalence under various future climate change scenarios. The data included in the analyses were collected from October 1, 2010, to April 30, 2018, and the current analyses were performed from January 2, 2022, to July 11, 2022. Exposure Annual ambient temperature exposure for each woman, estimated based on an atmospheric reanalysis model of the global climate. Main Outcomes and Measures The prevalence of IPV and its types (physical, sexual, and emotional violence) were assessed based on self-reported questionnaires from October 1, 2010, to April 30, 2018, and the changes in the prevalence with climate changes were estimated through the 2090s. Results The study included 194 871 ever-partnered women aged 15 to 49 years (mean [SD] age, 35.4 [7.6] years; overall IPV prevalence, 27.0%) from 3 South Asian countries. The prevalence of physical violence was highest (23.0%), followed by emotional (12.5%), and sexual violence (9.5%). The annual temperature ranges were mostly between 20 °C and 30 °C. A significant association was found between high ambient temperature and the prevalence of IPV against women, with each 1 °C increase in the annual mean temperature associated with a mean increase in IPV prevalence of 4.49% (95% CI, 4.20%-4.78%). According to the study's projections under the unlimited emissions scenarios (SSPs [shared socioeconomic pathways], as defined by the Intergovernmental Panel on Climate Change] 5-8.5), IPV prevalence would increase by 21.0% by the end of the 21st century, while it would only moderately increase under increasingly stricter scenarios (SSP2-4.5 [9.8%] and SSP1-2.6 [5.8%]). In addition, the projected increases in the prevalence of physical (28.3%) and sexual (26.1%) violence were greater than that of emotional violence (8.9%). In the 2090s, India was estimated to experience the highest IPV prevalence increase (23.5%) among the 3 countries, compared with Nepal (14.8%) and Pakistan (5.9%). Conclusions and Relevance This cross-sectional, multicountry study provides ample epidemiological evidence to support that high ambient temperature may be associated with the risk of IPV against women. These findings highlight the vulnerabilities and inequalities of women experiencing IPV in low- and middle-income countries in the context of global climate warming.
Collapse
Affiliation(s)
- Yixiang Zhu
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Cheng He
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Institute of Epidemiology, Neuherberg, Germany
| | - Michelle Bell
- School of Forestry and Environmental Studies, Yale University, New Haven, Connecticut
| | - Yuqiang Zhang
- Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Durham
| | - Zafar Fatmi
- Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | - Ying Zhang
- School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - Maryam Zaid
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Jovine Bachwenkizi
- Department of Environmental and Occupational Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Cong Liu
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
| | - Lu Zhou
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Renjie Chen
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
| | - Haidong Kan
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education, and National Health Commission Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
- IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
- Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| |
Collapse
|
43
|
Balapattabi K, Yavuz Y, Jiang J, Deng G, Mathieu NM, Ritter ML, Opichka MA, Reho JJ, McCorvy JD, Nakagawa P, Morselli LL, Mouradian GC, Atasoy D, Cui H, Hodges MR, Sigmund CD, Grobe JL. Angiotensin AT 1A receptor signal switching in Agouti-related peptide neurons mediates metabolic rate adaptation during obesity. Cell Rep 2023; 42:112935. [PMID: 37540598 PMCID: PMC10530419 DOI: 10.1016/j.celrep.2023.112935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/26/2023] [Accepted: 07/18/2023] [Indexed: 08/06/2023] Open
Abstract
Resting metabolic rate (RMR) adaptation occurs during obesity and is hypothesized to contribute to failed weight management. Angiotensin II (Ang-II) type 1 (AT1A) receptors in Agouti-related peptide (AgRP) neurons contribute to the integrative control of RMR, and deletion of AT1A from AgRP neurons causes RMR adaptation. Extracellular patch-clamp recordings identify distinct cellular responses of individual AgRP neurons from lean mice to Ang-II: no response, inhibition via AT1A and Gαi, or stimulation via Ang-II type 2 (AT2) receptors and Gαq. Following diet-induced obesity, a subset of Ang-II/AT1A-inhibited AgRP neurons undergo a spontaneous G-protein "signal switch," whereby AT1A stop inhibiting the cell via Gαi and instead begin stimulating the cell via Gαq. DREADD-mediated activation of Gαi, but not Gαq, in AT1A-expressing AgRP cells stimulates RMR in lean and obese mice. Thus, loss of AT1A-Gαi coupling within the AT1A-expressing AgRP neuron subtype represents a molecular mechanism contributing to RMR adaptation.
Collapse
Affiliation(s)
| | - Yavuz Yavuz
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Natalia M Mathieu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - McKenzie L Ritter
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Megan A Opichka
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John D McCorvy
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lisa L Morselli
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gary C Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
44
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
45
|
Suzuki M, Liu C, Oyama K, Yamazawa T. Trans-scale thermal signaling in biological systems. J Biochem 2023; 174:217-225. [PMID: 37461189 PMCID: PMC10464929 DOI: 10.1093/jb/mvad053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/21/2023] [Indexed: 08/31/2023] Open
Abstract
Biochemical reactions in cells serve as the endogenous source of heat, maintaining a constant body temperature. This process requires proper control; otherwise, serious consequences can arise due to the unwanted but unavoidable responses of biological systems to heat. This review aims to present a range of responses to heat in biological systems across various spatial scales. We begin by examining the impaired thermogenesis of malignant hyperthermia in model mice and skeletal muscle cells, demonstrating that the progression of this disease is caused by a positive feedback loop between thermally driven Ca2+ signaling and thermogenesis at the subcellular scale. After we explore thermally driven force generation in both muscle and non-muscle cells, we illustrate how in vitro assays using purified proteins can reveal the heat-responsive properties of proteins and protein assemblies. Building on these experimental findings, we propose the concept of 'trans-scale thermal signaling'.
Collapse
Key Words
- ATPase
- fluorescence microscopy
- heat-induced calcium release
- microheating
- type 1 ryanodine receptor.
Abbreviations: [Ca2+]i, intracellular Ca2+ concentration; CICR, Ca2+-induced Ca2+ release; ER, endoplasmic reticulum; FDB, flexor digitorum brevis; HEK293 cell, human embryonic kidney 293 cell; HICR, heat-induced Ca2+ release; IP3R, inositol 1,4,5-trisphosphate receptor; MH, malignant hyperthermia; RCC, rapid cooling contracture; RyR1, type 1 ryanodine receptor; SERCA, sarco/endoplasmic reticulum Ca2+-ATPase; SR, sarcoplasmic reticulum; TRP, transient receptor potential; WT, wild type
Collapse
Affiliation(s)
- Madoka Suzuki
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chujie Liu
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1, Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Kotaro Oyama
- Foundational Quantum Technology Research Directorate, National Institutes for Quantum Science and Technology, 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan
| | - Toshiko Yamazawa
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
46
|
Pongratz G, Straub RH. [Role of the sympathetic nervous system in chronic inflammation]. Z Rheumatol 2023:10.1007/s00393-023-01387-6. [PMID: 37488245 DOI: 10.1007/s00393-023-01387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 07/26/2023]
Abstract
In this review article the current model of the interaction between the sympathetic nervous system (SNS) and the immune system in the context of chronic inflammation is presented. Mechanisms in the interaction between the SNS and the immune system are shown, which are similar for all disease entities: 1) the biphasic effect of the sympathetic system on the inflammatory response with a proinflammatory, stimulating effect before and during the activation of the immune system (early) and a more inhibitory effect in late phases of immune activation (chronic). 2) The interruption of communication between immune cells and the brain by withdrawal of sympathetic nerve fibers from areas of inflammation, such as the spleen, lymph nodes or peripheral foci of inflammation. 3) The local replacement of catecholamines by neurotransmitter-producing cells to fine-tune the local immune response independently of the brain. 4) Increased activity of the SNS due to an imbalance of the autonomic nervous system at the systemic level, which provides an explanation for known disease sequelae and comorbidities due to the long duration of chronic inflammatory reactions, such as increased cardiovascular risk with hypertension, diabetes mellitus and catabolic metabolism. The understanding of neuroimmune interactions can lead to new therapeutic approaches, e.g., a stimulation of beta-adrenergic and even more an inhibition of alpha-adrenergic receptors or a restoration of the autonomic balance in the context of arthritis ) can make an anti-inflammatory contribution (more influence of the vagus nerve); however, in order to translate the theoretical findings into clinical action that is beneficial for the patient, controlled interventional studies are required.
Collapse
Affiliation(s)
- Georg Pongratz
- Abteilung für Rheumatologie und klinische Immunologie der Klinik für Gastroenterologie und interventionelle Endoskopie, Krankenhaus Barmherzige Brüder Regensburg, Prüfeninger Str. 86, 93049, Regensburg, Deutschland.
- Medizinische Fakultät, der Universität Regensburg, Regensburg, Deutschland.
| | - Rainer H Straub
- Labor für Experimentelle Rheumatologie und Neuroendokrino-Immunologie, Klinik und Poliklinik für Innere Medizin I, Universitätsklinikum, Regensburg, Deutschland
| |
Collapse
|
47
|
Yahiro T, Kataoka N, Nakamura K. Two Ascending Thermosensory Pathways from the Lateral Parabrachial Nucleus That Mediate Behavioral and Autonomous Thermoregulation. J Neurosci 2023; 43:5221-5240. [PMID: 37339876 PMCID: PMC10342230 DOI: 10.1523/jneurosci.0643-23.2023] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/03/2023] [Accepted: 06/10/2023] [Indexed: 06/22/2023] Open
Abstract
Thermoregulatory behavior in homeothermic animals is an innate behavior to defend body core temperature from environmental thermal challenges in coordination with autonomous thermoregulatory responses. In contrast to the progress in understanding the central mechanisms of autonomous thermoregulation, those of behavioral thermoregulation remain poorly understood. We have previously shown that the lateral parabrachial nucleus (LPB) mediates cutaneous thermosensory afferent signaling for thermoregulation. To understand the thermosensory neural network for behavioral thermoregulation, in the present study, we investigated the roles of ascending thermosensory pathways from the LPB in avoidance behavior from innocuous heat and cold in male rats. Neuronal tracing revealed two segregated groups of LPB neurons projecting to the median preoptic nucleus (MnPO), a thermoregulatory center (LPB→MnPO neurons), and those projecting to the central amygdaloid nucleus (CeA), a limbic emotion center (LPB→CeA neurons). While LPB→MnPO neurons include separate subgroups activated by heat or cold exposure of rats, LPB→CeA neurons were only activated by cold exposure. By selectively inhibiting LPB→MnPO or LPB→CeA neurons using tetanus toxin light chain or chemogenetic or optogenetic techniques, we found that LPB→MnPO transmission mediates heat avoidance, whereas LPB→CeA transmission contributes to cold avoidance. In vivo electrophysiological experiments showed that skin cooling-evoked thermogenesis in brown adipose tissue requires not only LPB→MnPO neurons but also LPB→CeA neurons, providing a novel insight into the central mechanism of autonomous thermoregulation. Our findings reveal an important framework of central thermosensory afferent pathways to coordinate behavioral and autonomous thermoregulation and to generate the emotions of thermal comfort and discomfort that drive thermoregulatory behavior.SIGNIFICANCE STATEMENT Coordination of behavioral and autonomous thermoregulation is important for maintaining thermal homeostasis in homeothermic animals. However, the central mechanism of thermoregulatory behaviors remains poorly understood. We have previously shown that the lateral parabrachial nucleus (LPB) mediates ascending thermosensory signaling that drives thermoregulatory behavior. In this study, we found that one pathway from the LPB to the median preoptic nucleus mediates heat avoidance, whereas the other pathway from the LPB to the central amygdaloid nucleus is required for cold avoidance. Surprisingly, both pathways are required for skin cooling-evoked thermogenesis in brown adipose tissue, an autonomous thermoregulatory response. This study provides a central thermosensory network that coordinates behavioral and autonomous thermoregulation and generates thermal comfort and discomfort that drive thermoregulatory behavior.
Collapse
Affiliation(s)
- Takaki Yahiro
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Naoya Kataoka
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
- Nagoya University Institute for Advanced Research, Nagoya, 464-8601, Japan
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
48
|
Rashid M, Kondoh K, Palfalvi G, Nakajima KI, Minokoshi Y. Inhibition of high-fat diet-induced inflammatory responses in adipose tissue by SF1-expressing neurons of the ventromedial hypothalamus. Cell Rep 2023; 42:112627. [PMID: 37339627 DOI: 10.1016/j.celrep.2023.112627] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/27/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023] Open
Abstract
Inflammation and thermogenesis in white adipose tissue (WAT) at different sites influence the overall effects of obesity on metabolic health. In mice fed a high-fat diet (HFD), inflammatory responses are less pronounced in inguinal WAT (ingWAT) than in epididymal WAT (epiWAT). Here we show that ablation and activation of steroidogenic factor 1 (SF1)-expressing neurons in the ventromedial hypothalamus (VMH) oppositely affect the expression of inflammation-related genes and the formation of crown-like structures by infiltrating macrophages in ingWAT, but not in epiWAT, of HFD-fed mice, with these effects being mediated by sympathetic nerves innervating ingWAT. In contrast, SF1 neurons of the VMH preferentially regulated the expression of thermogenesis-related genes in interscapular brown adipose tissue (BAT) of HFD-fed mice. These results suggest that SF1 neurons of the VMH differentially regulate inflammatory responses and thermogenesis among various adipose tissue depots and restrain inflammation associated with diet-induced obesity specifically in ingWAT.
Collapse
Affiliation(s)
- Misbah Rashid
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan.
| | - Gergo Palfalvi
- Division of Evolutionary Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Ken-Ichiro Nakajima
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|
49
|
Pongratz G, Straub RH. Chronic Effects of the Sympathetic Nervous System in Inflammatory Models. Neuroimmunomodulation 2023; 30:113-134. [PMID: 37231902 DOI: 10.1159/000530969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The immune system is embedded in a network of regulatory systems to keep homeostasis in case of an immunologic challenge. Neuroendocrine immunologic research has revealed several aspects of these interactions over the past decades, e.g., between the autonomic nervous system and the immune system. This review will focus on evidence revealing the role of the sympathetic nervous system (SNS) in chronic inflammation, like colitis, multiple sclerosis, systemic sclerosis, lupus erythematodes, and arthritis with a focus on animal models supported by human data. A theory of the contribution of the SNS in chronic inflammation will be presented that spans these disease entities. One major finding is the biphasic nature of the sympathetic contribution to inflammation, with proinflammatory effects until the point of disease outbreak and mainly anti-inflammatory influence thereafter. Since sympathetic nerve fibers are lost from sites of inflammation during inflammation, local cells and immune cells achieve the capability to endogenously produce catecholamines to fine-tune the inflammatory response independent of brain control. On a systemic level, it has been shown across models that the SNS is activated in inflammation as opposed to the parasympathetic nervous system. Permanent overactivity of the SNS contributes to many of the known disease sequelae. One goal of neuroendocrine immune research is defining new therapeutic targets. In this respect, it will be discussed that at least in arthritis, it might be beneficial to support β-adrenergic and inhibit α-adrenergic activity besides restoring autonomic balance. Overall, in the clinical setting, we now need controlled interventional studies to successfully translate the theoretical knowledge into benefits for patients.
Collapse
Affiliation(s)
- Georg Pongratz
- Department of Gastroenterology, Division of Rheumatology and Clinical Immunology, St. John of God Hospital, Regensburg, Germany
- Medical Faculty of the University of Regensburg, Regensburg, Germany
| | - Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrino-Immunology, Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
50
|
Janssen H, Kahles F, Liu D, Downey J, Koekkoek LL, Roudko V, D'Souza D, McAlpine CS, Halle L, Poller WC, Chan CT, He S, Mindur JE, Kiss MG, Singh S, Anzai A, Iwamoto Y, Kohler RH, Chetal K, Sadreyev RI, Weissleder R, Kim-Schulze S, Merad M, Nahrendorf M, Swirski FK. Monocytes re-enter the bone marrow during fasting and alter the host response to infection. Immunity 2023; 56:783-796.e7. [PMID: 36827982 PMCID: PMC10101885 DOI: 10.1016/j.immuni.2023.01.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/11/2022] [Accepted: 01/19/2023] [Indexed: 02/25/2023]
Abstract
Diet profoundly influences physiology. Whereas over-nutrition elevates risk for disease via its influence on immunity and metabolism, caloric restriction and fasting appear to be salutogenic. Despite multiple correlations observed between diet and health, the underlying biology remains unclear. Here, we identified a fasting-induced switch in leukocyte migration that prolongs monocyte lifespan and alters susceptibility to disease in mice. We show that fasting during the active phase induced the rapid return of monocytes from the blood to the bone marrow. Monocyte re-entry was orchestrated by hypothalamic-pituitary-adrenal (HPA) axis-dependent release of corticosterone, which augmented the CXCR4 chemokine receptor. Although the marrow is a safe haven for monocytes during nutrient scarcity, re-feeding prompted mobilization culminating in monocytosis of chronologically older and transcriptionally distinct monocytes. These shifts altered response to infection. Our study shows that diet-in particular, a diet's temporal dynamic balance-modulates monocyte lifespan with consequences for adaptation to external stressors.
Collapse
Affiliation(s)
- Henrike Janssen
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Florian Kahles
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dan Liu
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeffrey Downey
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura L Koekkoek
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vladimir Roudko
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darwin D'Souza
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lennard Halle
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wolfram C Poller
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher T Chan
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shun He
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - John E Mindur
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Máté G Kiss
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sumnima Singh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Atsushi Anzai
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kashish Chetal
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|