1
|
Li Y, Zhu Y, Liu Z, Ren R, Wang Y, Li Y, Zhang A, Xu H, Zhang Z, Tan Y, Ding Z, Huang Y. Acid-sensing ion channel 1a promotes LPS-induced acute lung injury through the circRNA 18-658/miR-127-5p/TRIM72 axis. Mol Immunol 2025; 182:150-159. [PMID: 40279987 DOI: 10.1016/j.molimm.2025.04.002] [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: 12/17/2024] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Acute lung injury (ALI) is a serious disease with sudden onset, rapid progression, poor treatment response and high mortality. Acid-sensitive ion channels (ASICs) are a type of cation channel activated by extracellular acidification and are involved in the pathogenesis of inflammatory and immune diseases. Our previous research revealed that ASIC1a promotes ALI, but the specific mechanism is unclear. Circular RNAs (circRNAs) are a large class of noncoding RNAs that play important roles in the pathological processes of many diseases. However, their role in lipopolysaccharide (LPS)-induced ALI and their correlation with the ASIC1a channel remain unclear. In this study, via high-throughput sequencing, we investigated the effect of ASIC1a on circRNA expression. We focused on circRNA18-658 and its downstream signaling pathway in rat models of ALI. Our results revealed that ASIC1a promotes ALI through the circRNA18-658/miR-127-5p/TRIM72 axis, providing new targets and ideas for the study of the mechanism of ALI and the development of new drugs.
Collapse
Affiliation(s)
- Yangyang Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yueqin Zhu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Zijun Liu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ruohan Ren
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yuyan Wang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yuemei Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Anqi Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hu Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ziwen Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yuanyuan Tan
- Emergency Department, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road. Hefei, Anhui 230022, China.
| | - Zhenxing Ding
- Emergency Department, the First Affiliated Hospital of Anhui Medical University, NO.218 Jixi Road. Hefei, Anhui 230022, China.
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmaceutical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
2
|
Schwarz KG, Pereyra KV, Díaz-Jara E, Vicencio SC, Del Rio R. Brainstem C1 neurons mediate heart failure decompensation and mortality during acute salt loading. Cardiovasc Res 2025; 121:241-253. [PMID: 39775485 PMCID: PMC12012444 DOI: 10.1093/cvr/cvae261] [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: 01/08/2024] [Revised: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
AIMS Heart failure (HF) is an emerging epidemic worldwide. Despite advances in treatment, the morbidity and mortality rate of HF remain high, and the global prevalence continues to rise. Common clinical features of HF include cardiac sympathoexcitation, disordered breathing, and kidney dysfunction; kidney dysfunction strongly contributes to sodium retention and fluid overload, leading to poor outcomes of HF patients. We have previously shown that brainstem pre-sympathetic neurons (C1) from the rostral ventrolateral medulla (RVLM) play a key role in sympathetic regulation in experimental models of HF. However, the role of RVLM-C1 neurons during salt-loading in the context of HF is unknown. This study tests whether RVLM C1 neurons drive cardiorespiratory decompensation and ultimately lead to sudden death in HF rats. METHODS AND RESULTS Adult male Sprague-Dawley rats underwent arteriovenous shunt to induce HF with preserved ejection fraction (HFpEF). Two weeks after HFpEF induction, bilateral selective ablation of RVLM C1 neurons was performed using anti-dopamine β-hydroxylase-saporin toxin. Animals were then fed a high Na+ diet (3% Na+ in food and 2% Na+ in water) for 3 weeks to induce compensated-to-decompensated HF state transition. Echocardiography, cardiac autonomic function, breathing function, and survival were assessed during the progression of HF. Salt loading resulted in marked decompensation in HF rats, as evidenced by a significant decrease in survival rates (survival: 10% vs. 100% HFpEF + Na+ vs. HFpEF). Furthermore, HFpEF + Na+ animals showed a further increase in cardiac sympathetic drive and more severe disordered breathing, including higher hypoxia-related epochs (i.e. apnoeas/hypopnoeas), compared with HF. Ablation of RVLM C1 neurons partly reduced the excessive cardiac sympathoexcitation during salt loading in HF, improved the exaggerated disordered breathing in HFpEF+ Na+ rats, and reduced decompensation-linked mortality. We found that hypoxia, but not high sodium, was the major contributor to impaired calcium handling in isolated adult cardiomyocytes. CONCLUSION Our results strongly suggest that RVLM C1 neurons contribute to acute HF decompensation during salt loading by a mechanism encompassing further increases in sympathetic outflow and hypoxia-related breathing disorders. This mechanism may ultimately impact cardiac contractility through cardiomyocyte calcium mishandling, increasing morbidity and mortality.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Sinay C Vicencio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Av. los Flamencos 01364, Punta Arenas 6210005, Chile
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd HLSIC-2091, Kansas City, KS 66160, USA
| |
Collapse
|
3
|
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
|
4
|
Hiyama TY. Understanding of Thirst in Medical Science. Yonago Acta Med 2025; 68:1-11. [PMID: 39968113 PMCID: PMC11831041 DOI: 10.33160/yam.2025.02.001] [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: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 02/20/2025]
Abstract
Thirst is fundamentally considered as a physiological function developed to maintain the homeostasis of body fluids. However, we occasionally experience thirst in situations that are not necessarily related to the maintenance of bodily fluid homeostasis. Because the only method available had been to quantify the degree of thirst using psychological indices, thirst research had made little progress until very recently. To quantitatively analyze thirst, it is necessary to elucidate the nature of the brain's "thirst center," which is believed to become active in response to thirst. Textbooks of physiology often refer to the "thirst center" in the hypothalamus, which is considered to sense the osmotic pressure of body fluids. However, they did not specify the location of this center in the hypothalamus. Furthermore, the existence of the so-called "osmotic pressure sensors" has yet to be confirmed. However, recently, a series of findings have been published that delve into the true nature of the thirst center. These advancements have been achieved using several new techniques, including the real-time monitoring of neural activities related to thirst regulation within the brains of experimental animals. At least at the animal level, recent advancements in experimental techniques have made it possible to objectively quantify the intensity of thirst as a physiological response. In this review article, the history of our research is presented and latest developments in thirst research are presented.
Collapse
Affiliation(s)
- Takeshi Y Hiyama
- Department of Integrative Physiology, Tottori University Graduate School and Faculty of Medicine, Tottori University, Yonago 683-8503, Japan and
- International Platform for Dryland Research and Education, Tottori University, Tottori 680-0001, Japan
| |
Collapse
|
5
|
Zhang Y, Ma K, Fang X, Zhang Y, Miao R, Guan H, Tian J. Targeting ion homeostasis in metabolic diseases: Molecular mechanisms and targeted therapies. Pharmacol Res 2025; 212:107579. [PMID: 39756557 DOI: 10.1016/j.phrs.2025.107579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/13/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
The incidence of metabolic diseases-hypertension, diabetes, obesity, metabolic dysfunction-associated steatotic liver disease (MASLD), and atherosclerosis-is increasing annually, imposing a significant burden on both human health and the social economy. The occurrence and development of these diseases are closely related to the disruption of ion homeostasis, which is crucial for maintaining cellular functions and metabolic equilibrium. However, the specific mechanism of ion homeostasis in metabolic diseases is still unclear. This article reviews the role of ion homeostasis in the pathogenesis of metabolic diseases and assesses its potential as a therapeutic target. Furthermore, the article explores pharmacological strategies that target ion channels and transporters, including existing drugs and emerging drugs under development. Lastly, the article discusses the development direction of future therapeutic strategies, including the possibility of gene therapy targeting specific ion channels and personalized therapy using novel biomarkers. In summary, targeting ion homeostasis provides a new perspective and potential therapeutic approach for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Kaile Ma
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huifang Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
6
|
Huang A, Yeum D, Sewaybricker LE, Aleksic S, Thomas M, Melhorn SJ, Earley YF, Schur EA. Update on Hypothalamic Inflammation and Gliosis: Expanding Evidence of Relevance Beyond Obesity. Curr Obes Rep 2025; 14:6. [PMID: 39775194 PMCID: PMC11963668 DOI: 10.1007/s13679-024-00595-8] [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] [Accepted: 11/14/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW To evaluate the role of hypothalamic inflammation and gliosis in human obesity pathogenesis and other disease processes influenced by obesity. RECENT FINDINGS Recent studies using established and novel magnetic resonance imaging (MRI) techniques to assess alterations in hypothalamic microarchitecture in humans support the presence of hypothalamic inflammation and gliosis in adults and children with obesity. Studies also identify prenatal exposure to maternal obesity or diabetes as a risk factor for hypothalamic inflammation and gliosis and increased obesity risk in offspring. Hypothalamic inflammation and gliosis have been further implicated in reproductive dysfunction (specifically polycystic ovarian syndrome and male hypogonadism), cardiovascular disease namely hypertension, and alterations in the gut microbiome, and may also accelerate neurocognitive aging. The most recent translational studies support the link between hypothalamic inflammation and gliosis and obesity pathogenesis in humans and expand our understanding of its influence on broader aspects of human health.
Collapse
Affiliation(s)
- Alyssa Huang
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Dabin Yeum
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Sandra Aleksic
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Melbin Thomas
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan J Melhorn
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Yumei Feng Earley
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Ellen A Schur
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Hönemann J, Hoffmann F, de Boni L, Gauger P, Mulder E, Möstl S, Heusser K, Schmitz M, Halbach M, Laurie SS, Lee SMC, Macias BR, Jordan J, Tank J. Impact of Daily Lower-Body Negative Pressure or Cycling Followed by Venous Constrictive Thigh Cuffs on Bedrest-Induced Orthostatic Intolerance. J Am Heart Assoc 2024; 13:e034800. [PMID: 39450723 PMCID: PMC11935683 DOI: 10.1161/jaha.124.034800] [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: 01/31/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Orthostatic intolerance occurs following immobilization in patients on Earth and in astronauts after spaceflight. Head-down tilt bedrest is a terrestrial model for weightlessness and induces orthostatic intolerance. We hypothesized that lower-body negative pressure (LBNP) or cycling followed by wearing venous constrictive thigh cuffs mitigates orthostatic intolerance after head-down tilt bedrest. METHODS AND RESULTS We enrolled 47 healthy individuals (20 women, 35±9 years) to a 30-day strict head-down tilt bedrest study. During bedrest, they were assigned to 6 hours of 25 mm Hg LBNP (n=12) per day and 1 hour of supine cycling followed by 6 hours of venous constriction through thigh cuffs 6 days per week (n=12), 6 hours of daily upright sitting (positive control, n=11), or no countermeasure (negative control, n=12). We measured orthostatic tolerance as the time to presyncope during 80° head-up tilt testing with incremental LBNP before and immediately after bedrest. We determined plasma volume with carbon monoxide rebreathing before and at the end of bedrest. After bedrest, orthostatic tolerance decreased 540±457 seconds in the control group, 539±68 seconds in the cycling group, 217±379 seconds in the LBNP group, and 289±89 seconds in the seated group (P<0.0001 time point, P=0.009 for group differences). Supine and upright heart rate increased in all groups following bedrest. Plasma volume was only maintained in the cycling group but decreased in all others (interaction countermeasure×time point P<0.0001). CONCLUSIONS Six hours of moderate LBNP training was as effective as sitting in attenuating orthostatic intolerance after 30 days of head-down tilt bedrest. Daily cycling exercise followed by 6 hours of wearing venous constrictive thigh cuffs, while maintaining plasma volume, did not improve orthostatic tolerance. REGISTRATION URL: https://www.bfarm.de/EN; Identifiers: DRKS00027643 and DRKS00030848.
Collapse
Affiliation(s)
- Jan‐Niklas Hönemann
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal MedicineUniversity of CologneCologneGermany
| | - Fabian Hoffmann
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal MedicineUniversity of CologneCologneGermany
| | - Laura de Boni
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
| | - Peter Gauger
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
| | - Edwin Mulder
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
| | - Stefan Möstl
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
| | - Karsten Heusser
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
| | - Marie‐Therese Schmitz
- Institute of Medical Biometry, Informatics and Epidemiology (IMBIE)University Hospital BonnBonnGermany
| | - Marcel Halbach
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal MedicineUniversity of CologneCologneGermany
| | | | | | | | - Jens Jordan
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
- Medical FacultyUniversity of CologneCologneGermany
| | - Jens Tank
- Institute of Aerospace MedicineGerman Aerospace CenterCologneGermany
| |
Collapse
|
8
|
Kubota H, Kunisawa K, Hasegawa M, Kurahashi H, Kagotani K, Fujimoto Y, Hayashi A, Sono R, Tsuji T, Saito K, Nabeshima T, Mouri A. Soy lysolecithin prevents hypertension and cognitive impairment induced in mice by high salt intake by inhibiting intestinal inflammation. Neurochem Int 2024; 180:105858. [PMID: 39271020 DOI: 10.1016/j.neuint.2024.105858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
High salt (HS) intake induces hypertension and cognitive impairment. Preventive strategies include against dietary supplements. Soybean lecithin is a widely used phospholipid supplement. Lysolecithin is important in cell signaling, digestion, and absorption. This study aimed to investigate the effects of lysophosphatidylcholine containing >70% of the total phospholipids (LPC70), on hypertension and cognitive impairment induced in mice by HS intake. Mice were provided with HS solution (2% NaCl in drinking water) with or without LPC70 for 12 weeks. Blood pressure, cognitive function, and inflammatory response of intestine were determined. Hypertension and impaired object recognition memory induced by HS intake were implicated with increased inducible nitric oxide synthase in the small intestine and tau hyperphosphorylation in the prefrontal cortex. LPC70 treatment prevented cognitive impairment by suppressing inducible nitric oxide synthase and tau hyperphosphorylation. LPC70 may be valuable as a functional food component in preventing HS-induced cognitive impairment.
Collapse
Affiliation(s)
- Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan; International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan; International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Hitomi Kurahashi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kazuhiro Kagotani
- Tsuji Oil Mills Co., Ltd, Mie, Japan; Tsuji Health & Beauty Science Laboratory, Mie University, Mie, Japan
| | | | | | | | | | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan; Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan; International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan; International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan.
| |
Collapse
|
9
|
Wan Q, Yang Z, Li L, Wu L. Central Angiotensin II type 1 receptor deficiency alleviates renal fibrosis by reducing sympathetic nerve discharge in nephrotoxic folic acid-induced chronic kidney disease. PeerJ 2024; 12:e18166. [PMID: 39346076 PMCID: PMC11439387 DOI: 10.7717/peerj.18166] [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: 05/23/2024] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Background Fibrosis after nephrotoxic injury is common. Activation of the paraventricular nucleus (PVN) renin-angiotensin system (RAS) and sympathetic nervous system (SNS) are common mechanism of renal fibrosis. However, there have limited knowledge about which brain regions are most affected by Angiotensin II (Ang II) after nephrotoxic injury, what role does Angiotensin II type 1a receptors (AT1R) signaling play and how this affects the outcomes of the kidneys. Methods In nephrotoxic folic acid-induced chronic kidney disease (FA-CKD) mouse models, we have integrated retrograde tracer techniques with studies on AT1afl/fl mice to pinpoint an excessively active central pathway that connects the paraventricular nucleus (PVN) to the rostral ventrolateral medulla (RVLM). This pathway plays a pivotal role in determining the kidney's fibrotic response following injury induced by folic acid. Results FA-CKD (vs sham) had increased in the kidney SNS activity and Ang II expression in the central PVN. The activation of Ang II in the PVN triggers the activation of the PVN-RVLM pathway, amplifies SNS output, thus facilitating fibrosis development in FA-CKD mouse. Blocking sympathetic traffic or deleting AT1a in the PVN alleviated renal fibrosis in FA-CKD mice. Conclusions The FA-CKD mice have increased the expression of Ang II in PVN, thereby activating AT1a-positive PVN neurons project to the RVLM, where SNS activity is engaged to initiate fibrotic processes. The Ang II in PVN may contribute to the development of kidney fibrosis after nephrotoxic folic acid-induced kidney injury.
Collapse
Affiliation(s)
- Qijun Wan
- Nephrology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Zhichen Yang
- Nephrology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Lingzhi Li
- Nephrology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Liling Wu
- Nephrology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Xia M, Wang T, Wang Y, Hu T, Chen D, Wang B. A neural perspective on the treatment of hypertension: the neurological network excitation and inhibition (E/I) imbalance in hypertension. Front Cardiovasc Med 2024; 11:1436059. [PMID: 39323755 PMCID: PMC11422145 DOI: 10.3389/fcvm.2024.1436059] [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: 05/21/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
Despite the increasing number of anti-hypertensive drugs have been developed and used in the clinical setting, persistent deficiencies persist, including issues such as lifelong dosage, combination therapy. Notwithstanding receiving the treatment under enduring these deficiencies, approximately 4 in 5 patients still fail to achieve reliable blood pressure (BP) control. The application of neuromodulation in the context of hypertension presents a pioneering strategy for addressing this condition, con-currently implying a potential central nervous mechanism underlying hypertension onset. We hypothesize that neurological networks, an essential component of maintaining appropriate neurological function, are involved in hypertension. Drawing on both peer-reviewed research and our laboratory investigations, we endeavor to investigate the underlying neural mechanisms involved in hypertension by identifying a close relationship between its onset of hypertension and an excitation and inhibition (E/I) imbalance. In addition to the involvement of excitatory glutamatergic and GABAergic inhibitory system, the pathogenesis of hypertension is also associated with Voltage-gated sodium channels (VGSCs, Nav)-mediated E/I balance. The overloading of glutamate or enhancement of glutamate receptors may be attributed to the E/I imbalance, ultimately triggering hypertension. GABA loss and GABA receptor dysfunction have also proven to be involved. Furthermore, we have identified that abnormalities in sodium channel expression and function alter neural excitability, thereby disturbing E/I balance and potentially serving as a mechanism underlying hypertension. These insights are expected to furnish potential strategies for the advancement of innovative anti-hypertensive therapies and a meaningful reference for the exploration of central nervous system (CNS) targets of anti-hypertensives.
Collapse
Affiliation(s)
- Min Xia
- Department of Anesthesiology, General Hospital of The Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, China
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Tianyu Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Yizhu Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Tingting Hu
- Department of Anesthesiology, General Hospital of The Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, China
| | - Defang Chen
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
- Emergency Intensive Care Unit, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| |
Collapse
|
11
|
Stock JM, Romberger NT, McMillan RK, Chung JW, Wenner MM, Stocker SD, Farquhar WB, Burciu RG. Acute hypernatremia increases functional connectivity of NaCl sensing regions in the human brain: An fMRI pilot study. Auton Neurosci 2024; 254:103182. [PMID: 38805791 PMCID: PMC12067945 DOI: 10.1016/j.autneu.2024.103182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Rodent studies demonstrated specialized sodium chloride (NaCl) sensing neurons in the circumventricular organs, which mediate changes in sympathetic nerve activity, arginine vasopressin, thirst, and blood pressure. However, the neural pathways involved in NaCl sensing in the human brain are incompletely understood. The purpose of this pilot study was to determine if acute hypernatremia alters the functional connectivity of NaCl-sensing regions of the brain in healthy young adults. Resting-state fMRI scans were acquired in 13 participants at baseline and during a 30 min hypertonic saline infusion (HSI). We used a seed-based approach to analyze the data, focusing on the subfornical organ (SFO) and the organum vasculosum of the lamina terminalis (OVLT) as regions of interest (ROIs). Blood chemistry and perceived thirst were assessed pre- and post-infusion. As expected, serum sodium increased from pre- to post-infusion in the HSI group. The primary finding of this pilot study was that the functional connectivity between the SFO and a cluster within the OVLT increased from baseline to the late-phase of the HSI. Bidirectional connectivity changes were found with cortical regions, with some regions showing increased connectivity with sodium-sensing regions while others showed decreased connectivity. Furthermore, the functional connectivity between the SFO and the posterior cingulate cortex (a control ROI) did not change from baseline to the late-phase of the HSI. This finding indicates a distinct response within the NaCl sensing network in the human brain specifically related to acute hypernatremia that will need to be replicated in large-scale studies.
Collapse
Affiliation(s)
- Joseph M Stock
- University of Delaware, Newark, DE, United States of America
| | | | | | - Jae Woo Chung
- University of Minnesota, Minneapolis, MN, United States of America
| | - Megan M Wenner
- University of Delaware, Newark, DE, United States of America
| | - Sean D Stocker
- University of Pittsburgh, Pittsburgh, PA, United States of America
| | | | - Roxana G Burciu
- University of Delaware, Newark, DE, United States of America.
| |
Collapse
|
12
|
Zhou S, Makashova O, Chevillard PM, Josey V, Li B, Prager-Khoutorsky M. Constitutive cell proliferation and neurogenesis in the organum vasculosum lamina terminalis and subfornical organ of adult rats. J Neuroendocrinol 2024; 36:e13377. [PMID: 38418229 DOI: 10.1111/jne.13377] [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: 10/14/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024]
Abstract
Neurogenesis continues throughout adulthood in the subventricular zone, hippocampal subgranular zone, and the hypothalamic median eminence (ME) and the adjacent medio-basal hypothalamus. The ME is one of the circumventricular organs (CVO), which are specialized brain areas characterized by an incomplete blood-brain barrier and, thus, are involved in mediating communication between the central nervous system and the periphery. Additional CVOs include the organum vasculosum laminae terminalis (OVLT) and the subfornical organs (SFO). Previous studies have demonstrated that the ME contains neural stem cells (NSCs) capable of generating new neurons and glia in the adult brain. However, it remains unclear whether the OVLT and SFO also contain proliferating cells, the identity of these cells, and their ability to differentiate into mature neurons. Here we show that glial and mural subtypes exhibit NSC characteristics, expressing the endogenous mitotic maker Ki67, and incorporating the exogenous mitotic marker BrdU in the OVLT and SFO of adult rats. Glial cells constitutively proliferating in the SFO comprise NG2 glia, while in the OVLT, both NG2 glia and tanycytes appear to constitute the NSC pool. Furthermore, pericytes, which are mural cells associated with capillaries, also contribute to the pool of cells constitutively proliferating in the OVLT and SFO of adult rats. In addition to these glial and mural cells, a fraction of NSCs containing proliferation markers Ki67 and BrdU also expresses the early postmitotic neuronal marker doublecortin, suggesting that these CVOs comprise newborn neurons. Notably, these neurons can differentiate and express the mature neuronal marker NeuN. These findings establish the sensory CVOs OVLT and SFO as additional neurogenic niches, where the generation of new neurons and glia persists in the adult brain.
Collapse
Affiliation(s)
- Suijian Zhou
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Olena Makashova
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Pierre-Marie Chevillard
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Vanessa Josey
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Banruo Li
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Masha Prager-Khoutorsky
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| |
Collapse
|
13
|
Stocker SD, Kinsman BJ, Farquhar WB, Gyarmati G, Peti-Peterdi J, Sved AF. Physiological Mechanisms of Dietary Salt Sensing in the Brain, Kidney, and Gastrointestinal Tract. Hypertension 2024; 81:447-455. [PMID: 37671571 PMCID: PMC10915107 DOI: 10.1161/hypertensionaha.123.19488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Excess dietary salt (NaCl) intake is strongly correlated with cardiovascular disease and is a major contributing factor to the pathogenesis of hypertension. NaCl-sensitive hypertension is a multisystem disorder that involves renal dysfunction, vascular abnormalities, and neurogenically-mediated increases in peripheral resistance. Despite a major research focus on organ systems and these effector mechanisms causing NaCl-induced increases in arterial blood pressure, relatively less research has been directed at elucidating how NaCl is sensed by various tissues to elicit these downstream effects. The purpose of this review is to discuss how the brain, kidney, and gastrointestinal tract sense NaCl including key cell types, the role of NaCl versus osmolality, and the underlying molecular and electrochemical mechanisms.
Collapse
Affiliation(s)
- Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine
| | - Brian J Kinsman
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital
| | | | - Georgina Gyarmati
- Department of Physiology and Neuroscience and Medicine, Zilkha Neurogenetic Institute, University of Southern California
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience and Medicine, Zilkha Neurogenetic Institute, University of Southern California
| | - Alan F. Sved
- Department of Neuroscience, University of Pittsburgh
| |
Collapse
|
14
|
Hiyama TY. Brain sodium sensing for regulation of thirst, salt appetite, and blood pressure. Physiol Rep 2024; 12:e15970. [PMID: 38479999 PMCID: PMC10937250 DOI: 10.14814/phy2.15970] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
The brain possesses intricate mechanisms for monitoring sodium (Na) levels in body fluids. During prolonged dehydration, the brain detects variations in body fluids and produces sensations of thirst and aversions to salty tastes. At the core of these processes Nax , the brain's Na sensor, exists. Specialized neural nuclei, namely the subfornical organ (SFO) and organum vasculosum of the lamina terminalis (OVLT), which lack the blood-brain barrier, play pivotal roles. Within the glia enveloping the neurons in these regions, Nax collaborates with Na+ /K+ -ATPase and glycolytic enzymes to drive glycolysis in response to elevated Na levels. Lactate released from these glia cells activates nearby inhibitory neurons. The SFO hosts distinct types of angiotensin II-sensitive neurons encoding thirst and salt appetite, respectively. During dehydration, Nax -activated inhibitory neurons suppress salt-appetite neuron's activity, whereas salt deficiency reduces thirst neuron's activity through cholecystokinin. Prolonged dehydration increases the Na sensitivity of Nax via increased endothelin expression in the SFO. So far, patients with essential hypernatremia have been reported to lose thirst and antidiuretic hormone release due to Nax -targeting autoantibodies. Inflammation in the SFO underlies the symptoms. Furthermore, Nax activation in the OVLT, driven by Na retention, stimulates the sympathetic nervous system via acid-sensing ion channels, contributing to a blood pressure elevation.
Collapse
Affiliation(s)
- Takeshi Y. Hiyama
- Department of Integrative PhysiologyTottori University Graduate School and Faculty of MedicineYonagoJapan
| |
Collapse
|
15
|
Rose CR, Verkhratsky A. Sodium homeostasis and signalling: The core and the hub of astrocyte function. Cell Calcium 2024; 117:102817. [PMID: 37979342 DOI: 10.1016/j.ceca.2023.102817] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/20/2023] [Indexed: 11/20/2023]
Abstract
Neuronal activity and neurochemical stimulation trigger spatio-temporal changes in the cytoplasmic concentration of Na+ ions in astrocytes. These changes constitute the substrate for Na+ signalling and are fundamental for astrocytic excitability. Astrocytic Na+ signals are generated by Na+ influx through neurotransmitter transporters, with primary contribution of glutamate transporters, and through cationic channels; whereas recovery from Na+ transients is mediated mainly by the plasmalemmal Na+/K+ ATPase. Astrocytic Na+ signals regulate the activity of plasmalemmal transporters critical for homeostatic function of astrocytes, thus providing real-time coordination between neuronal activity and astrocytic support.
Collapse
Affiliation(s)
- Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Alexej Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom; Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; International Collaborative Center on Big Science Plan for Purinergic Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| |
Collapse
|
16
|
Kajiwara S, Hasegawa Y, Fujimori K, Tomiyasu S, Kameno K, Uchikawa H, Morioka M. Persistent brain exposure to high sodium induces stroke onset by upregulation of cerebral microbleeds and oxidative stress in hypertensive rats. Hypertens Res 2024; 47:78-87. [PMID: 37783768 DOI: 10.1038/s41440-023-01447-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023]
Abstract
High salt intake induces hypertension and enhances stroke onset. However, whether an increase in brain sodium exposure itself is harmful and has poor prognosis remains unknown. Therefore, we employed hypertensive rats that underwent intracerebroventricular (ICV) infusion of sodium for 28 days and evaluated stroke onset and related cytotoxic brain injuries. Forty-seven spontaneously hypertensive stroke-prone (SHRSP) and 39 normotensive rats (Wistar Kyoto rats [WKY]) underwent persistent ICV infusion of the following four solutions: artificial cerebrospinal fluid, 0.9%, 2.7%, and 9% saline for 28 days. We evaluated stroke onset and all-cause mortality between SHRSP and WKY at each ICV sodium concentration as the primary endpoints. Our secondary objective was to explore histological brain injuries associated with SHRSP induced by high sodium ICV. The results indicated that ICV infusion of 2.7% and 9% sodium showed a significant increase in stroke onset, decrease in body weight, and increase rate of brain water content in SHRSP compared to WKY. Increased blood pressure was not observed for ICV infusion of high sodium, while serum sodium concentration was significantly increased in SHRSP compared to WKY. Histological evaluations revealed that higher sodium infusion significantly increased the number of activated microglia, superoxide, neuronal cell loss, and microbleeds compared to WKY and SHRSP with 0.9% sodium. We conclude that persistent exposure to high sodium in the brain is one of the risk factors for stroke onset upregulating cerebral microbleeds and oxidative stress in hypertensive rats.
Collapse
Affiliation(s)
- Sosho Kajiwara
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka, Japan
| | - Yu Hasegawa
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka, Japan.
- Department of Pharmaceutical Sciences, School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka, Japan.
| | - Kana Fujimori
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka, Japan
| | - Satoshi Tomiyasu
- Department of Medical Technology and Sciences, School of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan
| | - Koki Kameno
- Department of Neurosurgery, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Hiroki Uchikawa
- Department of Neurosurgery, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
17
|
Salgado-Mozo S, Thirouin ZS, Wyrosdic JC, García-Hernández U, Bourque CW. Na X Channel Is a Physiological [Na +] Detector in Oxytocin- and Vasopressin-Releasing Magnocellular Neurosecretory Cells of the Rat Supraoptic Nucleus. J Neurosci 2023; 43:8306-8316. [PMID: 37783507 PMCID: PMC10711705 DOI: 10.1523/jneurosci.1203-23.2023] [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: 06/29/2023] [Revised: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
The Scn7A gene encodes NaX, an atypical noninactivating Na+ channel, whose expression in sensory circumventricular organs is essential to maintain homeostatic responses for body fluid balance. However, NaX has also been detected in homeostatic effector neurons, such as vasopressin (VP)-releasing magnocellular neurosecretory cells (MNCVP) that secrete VP (antidiuretic hormone) into the bloodstream in response to hypertonicity and hypernatremia. Yet, the physiological relevance of NaX expression in these effector cells remains unclear. Here, we show that rat MNCVP in males and females is depolarized and excited in proportion with isosmotic increases in [Na+]. These responses were caused by an inward current resulting from a cell-autonomous increase in Na+ conductance. The Na+-evoked current was unaffected by blockers of other Na+-permeable ion channels but was significantly reduced by shRNA-mediated knockdown of Scn7A expression. Furthermore, reducing the density of NaX channels selectively impaired the activation of MNCVP by systemic hypernatremia without affecting their responsiveness to hypertonicity in vivo These results identify NaX as a physiological Na+ sensor, whose expression in MNCVP contributes to the generation of homeostatic responses to hypernatremia.SIGNIFICANCE STATEMENT In this study, we provide the first direct evidence showing that the sodium-sensing channel encoded by the Scn7A gene (NaX) mediates cell-autonomous sodium detection by MNCs in the low millimolar range and that selectively reducing the expression of these channels in MNCs impairs their activation in response to a physiologically relevant sodium stimulus in vitro and in vivo These data reveal that NaX operates as a sodium sensor in these cells and that the endogenous sensory properties of osmoregulatory effector neurons contribute to their homeostatic activation in vivo.
Collapse
Affiliation(s)
- Sandra Salgado-Mozo
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies, Instituto Politecnico Nacional, 07360 Mexico City, Mexico
| | - Zahra S Thirouin
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
| | - Joshua C Wyrosdic
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
| | - Ubaldo García-Hernández
- Department of Physiology, Biophysics and Neurosciences, Centre for Research and Advanced Studies, Instituto Politecnico Nacional, 07360 Mexico City, Mexico
| | - Charles W Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of McGill University Health Center, Montréal, Québec H3G1A4, Canada
| |
Collapse
|
18
|
Wang X, Wang H, Li J, Li L, Wang Y, Li A. Salt-induced phosphoproteomic changes in the subfornical organ in rats with chronic kidney disease. Ren Fail 2023; 45:2171886. [PMID: 36715439 PMCID: PMC9888458 DOI: 10.1080/0886022x.2023.2171886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVES Subfornical organ (SFO) is vital in chronic kidney disease (CKD) progression caused by high salt levels. The current study investigated the effects of high salt on phosphoproteomic changes in SFO in CKD rats. METHODS 5/6 nephrectomized rats were fed a normal-salt diet (0.4%) (NC group) or a high-salt diet (4%) (HC group) for three weeks, while sham-operated rats were fed a normal-salt diet (0.4%) (NS group). For phosphoproteomic analysis of SFO in different groups, TiO2 enrichment, isobaric tags for relative and absolute quantification (iTRAQ) labeling, and liquid chromatography-tandem mass spectrometry (LC-MS/MS) were used. RESULTS There were 6808 distinct phosphopeptides found, which corresponded to 2661 phosphoproteins. NC group had 168 upregulated and 250 downregulated phosphopeptides compared to NS group. Comparison to NC group, HC group had 154 upregulated and 124 downregulated phosphopeptides. Growth associated protein 43 (GAP43) and heat shock protein 27 (Hsp27) were significantly upregulated phosphoproteins and may protect against high-salt damage. Differential phosphoproteins with tight functional connection were synapse proteins and microtubule-associated proteins, implying that high-salt diet disrupted brain's structure and function. Furthermore, differential phosphoproteins in HC/NC comparison group were annotated to participate in GABAergic synapse signaling pathway and aldosterone synthesis and secretion, which attenuated inhibitory neurotransmitter effects and increased sympathetic nerve activity (SNA). DISCUSSION This large scale phosphoproteomic profiling of SFO sheds light on how salt aggravates CKD via the central nervous system.
Collapse
Affiliation(s)
- Xin Wang
- National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huizhen Wang
- National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawen Li
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lanying Li
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yifan Wang
- Anshun People’s Hospital of Guizhou Province, Anshun, China
| | - Aiqing Li
- National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China,CONTACT Aiqing Li National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
19
|
Wang J, Lv F, Yin W, Gao Z, Liu H, Wang Z, Sun J. The organum vasculosum of the lamina terminalis and subfornical organ: regulation of thirst. Front Neurosci 2023; 17:1223836. [PMID: 37732311 PMCID: PMC10507174 DOI: 10.3389/fnins.2023.1223836] [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: 05/17/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Thirst and water intake are regulated by the organum vasculosum of the lamina terminalis (OVLT) and subfornical organ (SFO), located around the anteroventral third ventricle, which plays a critical role in sensing dynamic changes in sodium and water balance in body fluids. Meanwhile, neural circuits involved in thirst regulation and intracellular mechanisms underlying the osmosensitive function of OVLT and SFO are reviewed. Having specific Nax channels in the glial cells and other channels (such as TRPV1 and TRPV4), the OVLT and SFO detect the increased Na+ concentration or hyperosmolality to orchestrate osmotic stimuli to the insular and cingulate cortex to evoke thirst. Meanwhile, the osmotic stimuli are relayed to the supraoptic nucleus (SON) and paraventricular nucleus of the hypothalamus (PVN) via direct neural projections or the median preoptic nucleus (MnPO) to promote the secretion of vasopressin which plays a vital role in the regulation of body fluid homeostasis. Importantly, the vital role of OVLT in sleep-arousal regulation is discussed, where vasopressin is proposed as the mediator in the regulation when OVLT senses osmotic stimuli.
Collapse
Affiliation(s)
- Jiaxu Wang
- Department of Anatomy and Neurobiology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Fenglin Lv
- Department of Anatomy and Neurobiology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Yin
- Department of Anatomy and Neurobiology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhanpeng Gao
- Department of Anatomy and Neurobiology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Hongyu Liu
- Institute of Sport and Exercise Medicine, North University of China, Taiyuan, China
| | - Zhen Wang
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhao Sun
- Department of Anatomy and Neurobiology, School of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
20
|
Xi H, Li X, Zhou Y, Sun Y. The Regulatory Effect of the Paraventricular Nucleus on Hypertension. Neuroendocrinology 2023; 114:1-13. [PMID: 37598678 DOI: 10.1159/000533691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Hypertension is among the most harmful factors of cardiovascular and cerebrovascular diseases and poses an urgent problem for the development of human society. In addition to previous studies on its pathogenesis focusing on the peripheral sympathetic nervous system, investigating the central causes of high blood pressure involving the neuroendocrine and neuroinflammatory mechanisms of the hypothalamic paraventricular nucleus (PVN) is paramount. This nucleus is considered to regulate the output of neurohormones and sympathetic nerve activity. In this article, we focussed on the neuroendocrine mechanism, primarily exploring the specific contributions and interactions of various neurons and neuroendocrine hormones, including GABAergic and glutamatergic neurons, nitric oxide, arginine vasopressin, oxytocin, and the renin-angiotensin system. Additionally, the neuroinflammatory mechanism in the PVN was discussed, encompassing microglia, reactive oxygen species, inflammatory factors, and pathways, as well as immune connections between the brain and extracerebral organs. Notably, the two central mechanisms involved in the PVN not only exist independently but also communicate with each other, jointly maintaining the hypertensive state of the body. Furthermore, we introduce well-known molecules and signal transduction pathways within the PVN that can play a regulatory role in the two mechanisms to provide a basis and inspire ideas for further research.
Collapse
Affiliation(s)
- Hanyu Xi
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xingru Li
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yun Zhou
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Department of Nephrology, Shanxi Provincial Integrated Traditional Chinese Medicine and Western Medicine Hospital, Taiyuan, China
| | - Yaojun Sun
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
21
|
Watso JC, Fancher IS, Gomez DH, Hutchison ZJ, Gutiérrez OM, Robinson AT. The damaging duo: Obesity and excess dietary salt contribute to hypertension and cardiovascular disease. Obes Rev 2023; 24:e13589. [PMID: 37336641 PMCID: PMC10406397 DOI: 10.1111/obr.13589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/08/2023] [Accepted: 05/24/2023] [Indexed: 06/21/2023]
Abstract
Hypertension is a primary risk factor for cardiovascular disease. Cardiovascular disease is the leading cause of death among adults worldwide. In this review, we focus on two of the most critical public health challenges that contribute to hypertension-obesity and excess dietary sodium from salt (i.e., sodium chloride). While the independent effects of these factors have been studied extensively, the interplay of obesity and excess salt overconsumption is not well understood. Here, we discuss both the independent and combined effects of excess obesity and dietary salt given their contributions to vascular dysfunction, autonomic cardiovascular dysregulation, kidney dysfunction, and insulin resistance. We discuss the role of ultra-processed foods-accounting for nearly 60% of energy intake in America-as a major contributor to both obesity and salt overconsumption. We highlight the influence of obesity on elevated blood pressure in the presence of a high-salt diet (i.e., salt sensitivity). Throughout the review, we highlight critical gaps in knowledge that should be filled to inform us of the prevention, management, treatment, and mitigation strategies for addressing these public health challenges.
Collapse
Affiliation(s)
- Joseph C. Watso
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Ibra S. Fancher
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, USA
| | - Dulce H. Gomez
- School of Kinesiology, Auburn University, Auburn, Alabama, USA
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - Orlando M. Gutiérrez
- Division of Nephrology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
22
|
Jin X, Xie J, Yeh CW, Chen JC, Cheng CJ, Lien CC, Huang CL. WNK1 promotes water homeostasis by acting as a central osmolality sensor for arginine vasopressin release. J Clin Invest 2023; 133:e164222. [PMID: 37071482 PMCID: PMC10231991 DOI: 10.1172/jci164222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 04/14/2023] [Indexed: 04/19/2023] Open
Abstract
Maintaining internal osmolality constancy is essential for life. Release of arginine vasopressin (AVP) in response to hyperosmolality is critical. Current hypotheses for osmolality sensors in circumventricular organs (CVOs) of the brain focus on mechanosensitive membrane proteins. The present study demonstrated that intracellular protein kinase WNK1 was involved. Focusing on vascular-organ-of-lamina-terminalis (OVLT) nuclei, we showed that WNK1 kinase was activated by water restriction. Neuron-specific conditional KO (cKO) of Wnk1 caused polyuria with decreased urine osmolality that persisted in water restriction and blunted water restriction-induced AVP release. Wnk1 cKO also blunted mannitol-induced AVP release but had no effect on osmotic thirst response. The role of WNK1 in the osmosensory neurons in CVOs was supported by neuronal pathway tracing. Hyperosmolality-induced increases in action potential firing in OVLT neurons was blunted by Wnk1 deletion or pharmacological WNK inhibitors. Knockdown of Kv3.1 channel in OVLT by shRNA reproduced the phenotypes. Thus, WNK1 in osmosensory neurons in CVOs detects extracellular hypertonicity and mediates the increase in AVP release by activating Kv3.1 and increasing action potential firing from osmosensory neurons.
Collapse
Affiliation(s)
- Xin Jin
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Jian Xie
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | | | - Jen-Chi Chen
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Chih-Jen Cheng
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Cheng-Chang Lien
- Institute of Neuroscience and
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chou-Long Huang
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
23
|
Cao W, Yang Z, Liu X, Ren S, Su H, Yang B, Liu Y, Wilcox CS, Hou FF. A kidney-brain neural circuit drives progressive kidney damage and heart failure. Signal Transduct Target Ther 2023; 8:184. [PMID: 37169751 PMCID: PMC10175540 DOI: 10.1038/s41392-023-01402-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 05/13/2023] Open
Abstract
Chronic kidney disease (CKD) and heart failure (HF) are highly prevalent, aggravate each other, and account for substantial mortality. However, the mechanisms underlying cardiorenal interaction and the role of kidney afferent nerves and their precise central pathway remain limited. Here, we combined virus tracing techniques with optogenetic techniques to map a polysynaptic central pathway linking kidney afferent nerves to subfornical organ (SFO) and thereby to paraventricular nucleus (PVN) and rostral ventrolateral medulla that modulates sympathetic outflow. This kidney-brain neural circuit was overactivated in mouse models of CKD or HF and subsequently enhanced the sympathetic discharge to both the kidney and the heart in each model. Interruption of the pathway by kidney deafferentation, selective deletion of angiotensin II type 1a receptor (AT1a) in SFO, or optogenetic silence of the kidney-SFO or SFO-PVN projection decreased the sympathetic discharge and lessened structural damage and dysfunction of both kidney and heart in models of CKD and HF. Thus, kidney afferent nerves activate a kidney-brain neural circuit in CKD and HF that drives the sympathetic nervous system to accelerate disease progression in both organs. These results demonstrate the crucial role of kidney afferent nerves and their central connections in engaging cardiorenal interactions under both physiological and disease conditions. This suggests novel therapies for CKD or HF targeting this kidney-brain neural circuit.
Collapse
Affiliation(s)
- Wei Cao
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Zhichen Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Xiaoting Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Siqiang Ren
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence; Key Laboratory of Mental Health of the Ministry of Education; Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, Guangdong, China
| | - Huanjuan Su
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Bihui Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University Medical Central, Washington, DC, USA
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China.
| |
Collapse
|
24
|
Leslie TK, Brackenbury WJ. Sodium channels and the ionic microenvironment of breast tumours. J Physiol 2023; 601:1543-1553. [PMID: 36183245 PMCID: PMC10953337 DOI: 10.1113/jp282306] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022] Open
Abstract
Cancers of epithelial origin such as breast, prostate, cervical, gastric, colon and lung cancer account for a large proportion of deaths worldwide. Better treatment of metastasis, the main cause of cancer deaths, is therefore urgently required. Several of these tumours have been shown to have an abnormally high concentration of Na+ ([Na+ ]) and emerging evidence points to this accumulation being due to elevated intracellular [Na+ ]. This poses intriguing questions about the cellular mechanisms underlying Na+ dysregulation in cancer, and its pathophysiological significance. Elevated intracellular [Na+ ] may be due to alterations in activity of the Na+ /K+ -ATPase, and/or increased influx via Na+ channels and Na+ -linked transporters. Maintenance of the electrochemical Na+ gradient across the plasma membrane is vital to power many cellular processes that are highly active in cancer cells, including glucose and glutamine import. Na+ channels are also upregulated in cancer cells, which in turn promotes tumour growth and metastasis. For example, ENaC and ASICs are overexpressed in cancers, increasing invasion and proliferation. In addition, voltage-gated Na+ channels are also upregulated in a range of tumour types, where they promote metastatic cell behaviours via various mechanisms, including membrane potential depolarisation and altered pH regulation. Together, recent findings relating to elevated Na+ in the tumour microenvironment and how this may be regulated by several classes of Na+ channels provide a link between altered Na+ handling and poor clinical outcome. There are new opportunities to leverage this altered Na+ microenvironment for therapeutic benefit, as exemplified by several ongoing clinical trials.
Collapse
Affiliation(s)
- Theresa K. Leslie
- Department of BiologyUniversity of YorkHeslingtonYorkUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkUK
| | - William J. Brackenbury
- Department of BiologyUniversity of YorkHeslingtonYorkUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkUK
| |
Collapse
|
25
|
Wei Y, Khalaf AT, Rui C, Abdul Kadir SY, Zainol J, Oglah Z. The Emergence of TRP Channels Interactome as a Potential Therapeutic Target in Pancreatic Ductal Adenocarcinoma. Biomedicines 2023; 11:biomedicines11041164. [PMID: 37189782 DOI: 10.3390/biomedicines11041164] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Integral membrane proteins, known as Transient Receptor Potential (TRP) channels, are cellular sensors for various physical and chemical stimuli in the nervous system, respiratory airways, colon, pancreas, bladder, skin, cardiovascular system, and eyes. TRP channels with nine subfamilies are classified by sequence similarity, resulting in this superfamily's tremendous physiological functional diversity. Pancreatic Ductal Adenocarcinoma (PDAC) is the most common and aggressive form of pancreatic cancer. Moreover, the development of effective treatment methods for pancreatic cancer has been hindered by the lack of understanding of the pathogenesis, partly due to the difficulty in studying human tissue samples. However, scientific research on this topic has witnessed steady development in the past few years in understanding the molecular mechanisms that underlie TRP channel disturbance. This brief review summarizes current knowledge of the molecular role of TRP channels in the development and progression of pancreatic ductal carcinoma to identify potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuanyuan Wei
- Basic Medical College, Chengdu University, Chengdu 610106, China
| | | | - Cao Rui
- Basic Medical College, Chengdu University, Chengdu 610106, China
| | - Samiah Yasmin Abdul Kadir
- Faculty of Medicine, Widad University College, BIM Point, Bandar Indera Mahkota, Kuantan 25200, Malaysia
| | - Jamaludin Zainol
- Faculty of Medicine, Widad University College, BIM Point, Bandar Indera Mahkota, Kuantan 25200, Malaysia
| | - Zahraa Oglah
- School of Science, Auckland University of Technology (AUT), 55 Wellesley Street, Auckland 1010, New Zealand
| |
Collapse
|
26
|
Stocker SD. Altered Neuronal Discharge in the Organum Vasculosum of the Lamina Terminalis Contributes to Dahl Salt-Sensitive Hypertension. Hypertension 2023; 80:872-881. [PMID: 36752103 PMCID: PMC10023399 DOI: 10.1161/hypertensionaha.122.20798] [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: 12/13/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND Salt-sensitive hypertension in humans and experimental models is associated with higher plasma and cerebrospinal fluid sodium chloride (NaCl) concentrations. Changes in extracellular NaCl concentrations are sensed by specialized neurons in the organum vasculosum of the lamina terminalis (OVLT). Stimulation of OVLT neurons increases sympathetic nerve activity (SNA) and arterial blood pressure (ABP), whereas chronic activation produces hypertension. Therefore, the present study tested whether OVLT neuronal activity was elevated and contributed to SNA and ABP in salt-sensitive hypertension. METHODS Male Dahl salt-sensitive (Dahl S) and Dahl salt-resistant (Dahl R) rats were fed 0.1% or 4.0% NaCl diets for 3 to 4 weeks and used for single-unit recordings of OVLT neurons or simultaneous recording of multiple sympathetic nerves during pharmacological inhibition of the OVLT. RESULTS Plasma and cerebrospinal fluid Na+ and Cl- concentrations were higher in Dahl S rats fed 4% versus 0.1% or Dahl R rats fed either diet. In vivo single-unit recordings revealed a significantly higher discharge of NaCl-responsive OVLT neurons in Dahl S rats fed 4% versus 0.1% or Dahl R rats. Interestingly, intracarotid infusion of hypertonic NaCl evoked greater increases in OVLT neuronal discharge of Dahl S versus Dahl R rats regardless of NaCl diet. The activity of non-NaCl-responsive OVLT neurons was not different across strain or diets. Finally, inhibition of OVLT neurons by local injection of the gamma-aminobutyric acid agonist muscimol produced a greater decrease in renal SNA, splanchnic SNA, and ABP of Dahl S rats fed 4% versus 0.1% or Dahl R rats. CONCLUSIONS A high salt diet activates NaCl-responsive OVLT neurons to increase SNA and ABP in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Sean D Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine, PA
| |
Collapse
|
27
|
Santos MM, Benvenuto GV, Ramos GV, Titotto AC, Adão MDS, de Lacerda LDCC, Lisbôa JAN, Lacerda-Neto JCD. Effect of Lactate Minimum Speed-Guided Conditioning on Selected Blood Parameters of Horses. J Equine Vet Sci 2022; 119:104133. [PMID: 36184020 DOI: 10.1016/j.jevs.2022.104133] [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: 02/21/2022] [Revised: 09/17/2022] [Accepted: 09/26/2022] [Indexed: 12/14/2022]
Abstract
During exercise, equines can suffer severe water and electrolyte imbalances depending on the intensity and duration. In this sense, conditioning aims to promote adaptations to the organism in order to maintain cardiovascular and thermoregulatory stability during exertion. This study aimed to evaluate the effect of conditioning guided by lactate minimum speed (LMS) test on the blood osmolality of horses. We hypothesized that after conditioning the blood osmolality would vary less during exercise and that LMS could be used in equine conditioning program. Ten Arabian horses were evaluated before (ET 1) and after (ET 2) 6 weeks of conditioning. The conditioning intensity was established from the LMS during ET 1. The blood was obtained at rest and during the ETs. An increase in LMS and a decrease in lactate were seen in individual horses; however, these differences were not significant at a group level. No change in blood osmolality was observed when comparing the ETs. The plasma volume remained unchanged in ET 2. The conditioning guided by LMS improved the animals' fitness, which was evidenced by the lower lactate production in ET 2. The fact that the osmolality kept unchanged proves the effectiveness of the osmotic blood balance during exercise, as its control involves the interaction of different systems. Body adaptations occurred with conditioning, providing greater homeostasis control since the plasma volume remained stable in ET 2. It was concluded that the LMS test can be used to define an effective equine conditioning program even though some adjustments are still necessary.
Collapse
Affiliation(s)
- Maíra Moreira Santos
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Guilherme Vechiato Benvenuto
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Gabriel Vieira Ramos
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Angélica Cristina Titotto
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Milena Dos Santos Adão
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Luciana de Cenço Corrêa de Lacerda
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil
| | | | - José Corrêa de Lacerda-Neto
- Department of Clinic and Veterinary Surgery, Faculty of Agricultural and Veterinary Sciences, UNESP, São Paulo State University, Jaboticabal, São Paulo, Brazil.
| |
Collapse
|
28
|
Sudbury JR, Zaelzer C, Trudel E, Bumagin A, Bourque CW. Synaptic control of rat magnocellular neurosecretory cells by warm-sensing neurons in the organum vasculosum lamina terminalis. J Neuroendocrinol 2022; 34:e13214. [PMID: 36426844 DOI: 10.1111/jne.13214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 10/07/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Abstract
Increases in core body temperature cause secretion of vasopressin (vasopressin, antidiuretic hormone) to promote water reabsorption and blunt water losses incurred through homeostatic evaporative cooling. Subtypes of transient receptor potential vanilloid (Trpv) channels have been shown to contribute to the intrinsic regulation of vasopressin-releasing magnocellular neurosecretory cells (MNCs) in the supraoptic nucleus (SON) and paraventricular nucleus (PVN). However, MNCs in vivo can also be excited by local heating of the adjacent preoptic area, indicating they receive thermosensory information from other areas. Here, we investigated whether neurons in the organum vasculosum lamina terminalis (OVLT) contribute to this process using in vitro electrophysiological approaches in male rats. We found that the majority of OVLT neurons are thermosensitive in the physiological range (36-39°C) and that this property is retained under conditions blocking synaptic transmission. A subset of these neurons could be antidromically activated by electrical stimulation in the SON. Whole cell recordings from SON MNCs revealed that heating significantly increases the rate of spontaneous excitatory postsynaptic currents (sEPCSs), and that this response is abolished by lesions targeting the OVLT, but not by bilateral lesions placed in the adjacent preoptic area. Finally, local heating of the OVLT caused a significant excitation of MNCs in the absence of temperature changes in the SON, and this effect was blocked by inhibitors of ionotropic glutamate receptors. These findings indicate that the OVLT serves as an important thermosensory nucleus and contributes to the activation of MNCs during physiological heating.
Collapse
Affiliation(s)
- Jessica R Sudbury
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Cristian Zaelzer
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Eric Trudel
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Anna Bumagin
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Charles W Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
29
|
The Central Nervous Mechanism of Stress-Promoting Cancer Progression. Int J Mol Sci 2022; 23:ijms232012653. [PMID: 36293510 PMCID: PMC9604265 DOI: 10.3390/ijms232012653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022] Open
Abstract
Evidence shows that stress can promote the occurrence and development of tumors. In recent years, many studies have shown that stress-related hormones or peripheral neurotransmitters can promote the proliferation, survival, and angiogenesis of tumor cells and impair the body’s immune response, causing tumor cells to escape the “surveillance” of the immune system. However, the perception of stress occurs in the central nervous system (CNS) and the role of the central nervous system in tumor progression is still unclear, as are the underlying mechanisms. This review summarizes what is known of stress-related CNS-network activation during the stress response and the influence of the CNS on tumors and discusses available adjuvant treatment methods for cancer patients with negative emotional states, such as anxiety and depression.
Collapse
|
30
|
Wang W, Xu M, Yue J, Zhang Q, Nie X, Jin Y, Zhang Z. Knockdown of Acid-sensing Ion Channel 1a in the PVN Promotes Metabolic Disturbances in Male Mice. Endocrinology 2022; 163:6650558. [PMID: 35894166 DOI: 10.1210/endocr/bqac115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Indexed: 11/19/2022]
Abstract
Increasing incidence of metabolic disturbances has become a severe public healthcare problem. Ion channels and receptors in the paraventricular nucleus (PVN) of the hypothalamus serve vital roles in modulating neuronal activities and endocrine functions, which are linked to the regulation of energy balance and glucose metabolism. In this study, we found that acid-sensing ion channel 1a (ASIC1a), a Ca2+-permeable cationic ion channel was localized in the PVN. Knockdown of ASIC1a in this region led to significant body weight gain, glucose intolerance, and insulin resistance. Pharmacological inhibition of ASIC1a resulted in an increase in food intake and a decrease in energy expenditure. Our findings suggest ASIC1a in the PVN as a potential new target for the therapeutic intervention of metabolic disorders.
Collapse
Affiliation(s)
- Wei Wang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengyun Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiayin Yue
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qilun Zhang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaomin Nie
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Jin
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
31
|
ASIC1a induces mitochondrial apoptotic responses in acute lung injury. Eur J Pharmacol 2022; 934:175296. [PMID: 36162458 DOI: 10.1016/j.ejphar.2022.175296] [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: 07/05/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/20/2022]
Abstract
AIM This study aimed to investigate the promoting effect of acid-sensing ion channel 1a (ASIC1a) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and its mechanisms. METHODS In this experiment, the ALI rat model was induced by intratracheal injection of LPS, and the ASIC1a specific blocker psalmotoxin-1 (PcTx-1) was injected into the tail vein before LPS administration once. Western blot, immunofluorescence, immunohistochemistry and real-time PCR methods were used to detect ASIC1a and apoptosis-related proteins expressions in lung tissue and RLE-6TN rat type II alveolar epithelial cells. Confocal Laser Scanning Microscopy was used to detect Ca2+ fluorescence intensity in RLE-6TN cells. RESULTS PcTx-1 pretreatment not only inhibited the pathological changes of LPS-induced ALI in lung tissue, but also inhibited lung dysfunction. PcTx-1 also reduced the increased levels of the apoptosis-related proteins B-cell lymphoma-2-associated X (Bax) and cleaved cysteinyl aspartate specific proteinase 3 (Cleaved caspase-3) and increased the decreased level of B-cell lymphoma-2 (Bcl-2) in the lung tissue of the model group. LPS-induced changes in mitochondrial membrane potential and calcium influx in alveolar epithelial cells were also reversed by PcTx-1. CONCLUSION ASIC1a induces an apoptotic response in ALI through mitochondrial apoptosis.
Collapse
|
32
|
Ohara H, Nabika T. Genetic Modifications to Alter Blood Pressure Level. Biomedicines 2022; 10:biomedicines10081855. [PMID: 36009402 PMCID: PMC9405136 DOI: 10.3390/biomedicines10081855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 12/04/2022] Open
Abstract
Genetic manipulation is one of the indispensable techniques to examine gene functions both in vitro and in vivo. In particular, cardiovascular phenotypes such as blood pressure cannot be evaluated in vitro system, necessitating the creation of transgenic or gene-targeted knock-out and knock-in experimental animals to understand the pathophysiological roles of specific genes on the disease conditions. Although genome-wide association studies (GWAS) in various human populations have identified multiple genetic variations associated with increased risk for hypertension and/or its complications, the causal links remain unresolved. Genome-editing technologies can be applied to many different types of cells and organisms for creation of knock-out/knock-in models. In the post-GWAS era, it may be more worthwhile to validate pathophysiological implications of the risk variants and/or candidate genes by creating genome-edited organisms.
Collapse
|
33
|
Luan SH, Yang YQ, Ye MP, Liu H, Rao QF, Kong JL, Wu FR. ASIC1a promotes hepatic stellate cell activation through the exosomal miR-301a-3p/BTG1 pathway. Int J Biol Macromol 2022; 211:128-139. [PMID: 35561854 DOI: 10.1016/j.ijbiomac.2022.05.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022]
Abstract
Activation of hepatic stellate cells (HSCs) is a key cause of liver fibrosis. However, the mechanisms leading to the activation of HSCs are not fully understood. In the pathological process, acid-sensing ion channel 1a (ASIC1a) is widely involved in the development of inflammatory diseases, suggesting that ASIC1a may play an important role in liver fibrosis. We found that in an acidic environment, ASIC1a leads to HSC-T6 cell activation. Meanwhile, exosomes produced by activated HSC-T6 cells (HSC-EXOs) can be reabsorbed by quiescent HSC-T6 cells to promote their activation. Exosomes mainly carry miRNAs involved in intercellular information exchange. We performed exosome miRNA whole transcriptome sequencing. The results indicated that the acidic environment could alter the miRNA expression profile in the exosomes of HSC-T6 cells. Further studies revealed that ASIC1a promotes the activation of HSCs by regulating miR-301a-3p targeting B-cell translocation gene 1 (BTG1). In conclusion, our study found that ASIC1a may affect HSC activation through the exosomal miR-301a-3p/BTG1 axis, and inhibiting ASIC1a may be a promising treatment strategy for liver fibrosis.
Collapse
Affiliation(s)
- Shao-Hua Luan
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | | | - Man-Ping Ye
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Hui Liu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Qiu-Fan Rao
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Jin-Ling Kong
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Fan-Rong Wu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China.
| |
Collapse
|
34
|
TRPV1-Mediated Sensing of Sodium and Osmotic Pressure in POMC Neurons in the Arcuate Nucleus of the Hypothalamus. Nutrients 2022; 14:nu14132600. [PMID: 35807782 PMCID: PMC9268643 DOI: 10.3390/nu14132600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/11/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The central melanocortin system conducted by anorexigenic pro-opiomelanocortin (POMC) neurons and orexigenic agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus (ARC) not only regulates feeding behavior but also blood pressure. Excessive salt intake raises the Na+ concentration ([Na+]) in the cerebrospinal fluid (CSF) and worsens hypertension. The blood–brain barrier is immature in the ARC. Therefore, both AgRP and POMC neurons in the ARC have easy access to the electrolytes in the blood and can sense changes in their concentrations. However, the sensitivity of AgRP and POMC neurons to Na+ remains unclear. This study aimed to explore how the changes in the extracellular Na+ concentration ([Na+]) influence these neurons by measuring the cytosolic Ca2+ concentration ([Ca2+]i) in the single neurons isolated from the ARC that were subsequently immunocytochemically identified as AgRP or POMC neurons. Both AgRP and POMC neurons responded to increases in both [Na+] and osmolarity in C57BL/6 mice. In contrast, in transient receptor potential vanilloid 1 (TRPV1) knockout (KO) mice, POMC neurons failed to respond to increases in both [Na+] and osmolarity, while they responded to high glucose and angiotensin II levels with increases in [Ca2+]i. Moreover, in KO mice fed a high-salt diet, the expression of POMC was lower than that in wild-type mice. These results demonstrate that changes in [Na+] and osmolarity are sensed by the ARC POMC neurons via the TRPV1-dependent mechanism.
Collapse
|
35
|
Davis H, Paterson DJ, Herring N. Post-Ganglionic Sympathetic Neurons can Directly Sense Raised Extracellular Na + via SCN7a/Na x. Front Physiol 2022; 13:931094. [PMID: 35784866 PMCID: PMC9247455 DOI: 10.3389/fphys.2022.931094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
The relationship between dietary NaCl intake and high blood pressure is well-established, and occurs primarily through activation of the sympathetic nervous system. Nax, a Na+-sensitive Na+ channel, plays a pivotal role in driving sympathetic excitability, which is thought to originate from central regions controlling neural outflow. We investigated whether post-ganglionic sympathetic neurons from different ganglia innervating cardiac and vasculature tissue can also directly sense extracellular Na+. Using whole-cell patch clamp recordings we demonstrate that sympathetic neurons from three sympathetic ganglia (superior cervical, stellate and superior mesenteric/coeliac) respond to elevated extracellular NaCl concentration. In sympathetic stellate ganglia neurons, we established that the effect of NaCl was dose-dependent and independent of osmolarity, Cl- and membrane Ca2+ flux, and critically dependent on extracellular Na+ concentration. We show that Nax is expressed in sympathetic stellate ganglia neurons at a transcript and protein level using single-cell RNA-sequencing and immunohistochemistry respectively. Additionally, the response to NaCl was prevented by siRNA-mediated knockdown of Nax, but not by inhibition of other membrane Na+ pathways. Together, these results demonstrate that post-ganglionic sympathetic neurons are direct sensors of extracellular Na+ via Nax, which could contribute to sympathetic driven hypertension.
Collapse
Affiliation(s)
- Harvey Davis
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Wellcome Trust OXION Initiative in Ion Channels and Disease, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Wellcome Trust OXION Initiative in Ion Channels and Disease, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
36
|
Li Y, Cui B, Zhang S, Li B, Li J, Liu S, Zhao Q. Ion-Selective Organic Electrochemical Transistors: Recent Progress and Challenges. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107413. [PMID: 35182018 DOI: 10.1002/smll.202107413] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Indexed: 06/14/2023]
Abstract
The charged species inside biofluids (blood, interstitial fluid, sweat, saliva, urine, etc.) can reflect the human body's physiological conditions and thus be adopted to diagnose various diseases early. Among all personalized health management applications, ion-selective organic electrochemical transistors (IS-OECTs) have shown tremendous potential in point-of-care testing of biofluids due to low cost, ease of fabrication, high signal amplification, and low detection limit. Moreover, IS-OECTs exhibit excellent flexibility and biocompatibility that enable their application in wearable bioelectronics for continuous health monitoring. In this review, the working principle of IS-OECTs and the recent studies of IS-OECTs for performance improvement are reviewed. Specifically, contemporary studies on material design and device optimization to enhance the sensitivity of IS-OECTs are discussed. In addition, the progress toward the commercialization of IS-OECTs is highlighted, and the recently proposed solutions or alternatives are summarized. The main challenges and perspectives for fully exploiting IS-OECTs toward future preventive and personal medical devices are addressed.
Collapse
Affiliation(s)
- Yang Li
- College of Electronic and Optical Engineering, and College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NJUPT), 9 Wenyuan Road, Nanjing, Jiangsu, 210023, China
| | - Binbin Cui
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Shiming Zhang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Bingxiang Li
- College of Electronic and Optical Engineering, and College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NJUPT), 9 Wenyuan Road, Nanjing, Jiangsu, 210023, China
| | - Jianmin Li
- College of Electronic and Optical Engineering, and College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NJUPT), 9 Wenyuan Road, Nanjing, Jiangsu, 210023, China
| | - Shujuan Liu
- State Key Laboratory of Organic Electronics and Information Displays, and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NJUPT), 9 Wenyuan Road, Nanjing, Jiangsu, 210023, China
| | - Qiang Zhao
- College of Electronic and Optical Engineering, and College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications (NJUPT), 9 Wenyuan Road, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Organic Electronics and Information Displays, and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NJUPT), 9 Wenyuan Road, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
37
|
Structure-guided unlocking of Na X reveals a non-selective tetrodotoxin-sensitive cation channel. Nat Commun 2022; 13:1416. [PMID: 35301303 PMCID: PMC8931054 DOI: 10.1038/s41467-022-28984-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
Unlike classical voltage-gated sodium (NaV) channels, NaX has been characterized as a voltage-insensitive, tetrodotoxin-resistant, sodium (Na+)-activated channel involved in regulating Na+ homeostasis. However, NaX remains refractory to functional characterization in traditional heterologous systems. Here, to gain insight into its atypical physiology, we determine structures of the human NaX channel in complex with the auxiliary β3-subunit. NaX reveals structural alterations within the selectivity filter, voltage sensor-like domains, and pore module. We do not identify an extracellular Na+-sensor or any evidence for a Na+-based activation mechanism in NaX. Instead, the S6-gate remains closed, membrane lipids fill the central cavity, and the domain III-IV linker restricts S6-dilation. We use protein engineering to identify three pore-wetting mutations targeting the hydrophobic S6-gate that unlock a robust voltage-insensitive leak conductance. This constitutively active NaX-QTT channel construct is non-selective among monovalent cations, inhibited by extracellular calcium, and sensitive to classical NaV channel blockers, including tetrodotoxin. Our findings highlight a functional diversity across the NaV channel scaffold, reshape our understanding of NaX physiology, and provide a template to demystify recalcitrant ion channels.
Collapse
|
38
|
Stock JM, Chelimsky G, Edwards DG, Farquhar WB. Dietary sodium and health: How much is too much for those with orthostatic disorders? Auton Neurosci 2022; 238:102947. [PMID: 35131651 PMCID: PMC9296699 DOI: 10.1016/j.autneu.2022.102947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/09/2021] [Accepted: 01/16/2022] [Indexed: 10/19/2022]
Abstract
High dietary salt (NaCl) increases blood pressure (BP) and can adversely impact multiple target organs including the vasculature, heart, kidneys, brain, autonomic nervous system, skin, eyes, and bone. However, patients with orthostatic disorders are told to increase their NaCl intake to help alleviate symptoms. While there is evidence to support the short-term benefits of increasing NaCl intake in these patients, there are few studies assessing the benefits and side effects of long-term high dietary NaCl. The evidence reviewed suggests that high NaCl can adversely impact multiple target organs, often independent of BP. However, few of these studies have been performed in patients with orthostatic disorders. We conclude that the recommendation to increase dietary NaCl in patients with orthostatic disorders should be done with care, keeping in mind the adverse impact on dietary NaCl in people without orthostatic disorders. Modest, rather than robust, increases in NaCl intake may be sufficient to alleviate symptoms but also minimize any long-term negative effects.
Collapse
Affiliation(s)
- Joseph M Stock
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States of America
| | - Gisela Chelimsky
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States of America
| | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States of America.
| |
Collapse
|
39
|
Hunter RW, Dhaun N, Bailey MA. The impact of excessive salt intake on human health. Nat Rev Nephrol 2022; 18:321-335. [DOI: 10.1038/s41581-021-00533-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2021] [Indexed: 12/19/2022]
|
40
|
NODA M, MATSUDA T. Central regulation of body fluid homeostasis. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2022; 98:283-324. [PMID: 35908954 PMCID: PMC9363595 DOI: 10.2183/pjab.98.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Extracellular fluids, including blood, lymphatic fluid, and cerebrospinal fluid, are collectively called body fluids. The Na+ concentration ([Na+]) in body fluids is maintained at 135-145 mM and is broadly conserved among terrestrial animals. Homeostatic osmoregulation by Na+ is vital for life because severe hyper- or hypotonicity elicits irreversible organ damage and lethal neurological trauma. To achieve "body fluid homeostasis" or "Na homeostasis", the brain continuously monitors [Na+] in body fluids and controls water/salt intake and water/salt excretion by the kidneys. These physiological functions are primarily regulated based on information on [Na+] and relevant circulating hormones, such as angiotensin II, aldosterone, and vasopressin. In this review, we discuss sensing mechanisms for [Na+] and hormones in the brain that control water/salt intake behaviors, together with the responsible sensors (receptors) and relevant neural pathways. We also describe mechanisms in the brain by which [Na+] increases in body fluids activate the sympathetic neural activity leading to hypertension.
Collapse
Affiliation(s)
- Masaharu NODA
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
- Correspondence should be addressed to: Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama, Kanagawa 226-8503, Japan (e-mail: )
| | - Takashi MATSUDA
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
41
|
Affiliation(s)
- David H Ellison
- From the Oregon Clinical and Translational Research Institute, Oregon Health and Science University (D.H.E) and the VA Portland Health Care System (D.H.E.) - both in Portland; and LeDucq Transatlantic Network of Excellence (D.H.E., P.W.) and the Departments of Medicine and Physiology, Johns Hopkins University (P.W.) - both in Baltimore
| | - Paul Welling
- From the Oregon Clinical and Translational Research Institute, Oregon Health and Science University (D.H.E) and the VA Portland Health Care System (D.H.E.) - both in Portland; and LeDucq Transatlantic Network of Excellence (D.H.E., P.W.) and the Departments of Medicine and Physiology, Johns Hopkins University (P.W.) - both in Baltimore
| |
Collapse
|
42
|
Yamamoto A, Otani K, Okada M, Yamawaki H. Chemokine-like Receptor 1 in Brain of Spontaneously Hypertensive Rats Mediates Systemic Hypertension. Int J Mol Sci 2021; 22:11812. [PMID: 34769243 PMCID: PMC8584015 DOI: 10.3390/ijms222111812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022] Open
Abstract
Adipocytokine chemerin is a biologically active molecule secreted from adipose tissue. Chemerin elicits a variety of functions via chemokine-like receptor 1 (CMKLR1). The cardiovascular center in brain that regulates blood pressure (BP) is involved in pathophysiology of systemic hypertension. Thus, we explored the roles of brain chemerin/CMKLR1 on regulation of BP in spontaneously hypertensive rats (SHR). For this aim, we examined effects of intracerebroventricular (i.c.v.) injection of CMKLR1 small interfering (si)RNA on both systemic BP as measured by tail cuff system and protein expression in paraventricular nucleus (PVN) of SHR as determined by Western blotting. We also examined both central and peripheral protein expression of chemerin by Western blotting. Systolic BP of SHR but not normotensive Wistar Kyoto rats (WKY) was decreased by CMKLR1 siRNA. The decrease of BP by CMKLR1 siRNA persisted for 3 days. Protein expression of CMKLR1 in PVN of SHR tended to be increased compared with WKY, which was suppressed by CMKLR1 siRNA. Protein expression of chemerin in brain, peripheral plasma, and adipose tissue was not different between WKY and SHR. In summary, we for the first time revealed that the increased protein expression of CMKLR1 in PVN is at least partly responsible for systemic hypertension in SHR.
Collapse
Affiliation(s)
| | | | | | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada, Aomori 034-8628, Japan; (A.Y.); (K.O.); (M.O.)
| |
Collapse
|
43
|
Jeong JK, Dow SA, Young CN. Sensory Circumventricular Organs, Neuroendocrine Control, and Metabolic Regulation. Metabolites 2021; 11:metabo11080494. [PMID: 34436435 PMCID: PMC8402088 DOI: 10.3390/metabo11080494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022] Open
Abstract
The central nervous system is critical in metabolic regulation, and accumulating evidence points to a distributed network of brain regions involved in energy homeostasis. This is accomplished, in part, by integrating peripheral and central metabolic information and subsequently modulating neuroendocrine outputs through the paraventricular and supraoptic nucleus of the hypothalamus. However, these hypothalamic nuclei are generally protected by a blood-brain-barrier limiting their ability to directly sense circulating metabolic signals—pointing to possible involvement of upstream brain nuclei. In this regard, sensory circumventricular organs (CVOs), brain sites traditionally recognized in thirst/fluid and cardiovascular regulation, are emerging as potential sites through which circulating metabolic substances influence neuroendocrine control. The sensory CVOs, including the subfornical organ, organum vasculosum of the lamina terminalis, and area postrema, are located outside the blood-brain-barrier, possess cellular machinery to sense the metabolic interior milieu, and establish complex neural networks to hypothalamic neuroendocrine nuclei. Here, evidence for a potential role of sensory CVO-hypothalamic neuroendocrine networks in energy homeostasis is presented.
Collapse
Affiliation(s)
| | | | - Colin N. Young
- Correspondence: ; Tel.: +1-202-994-9575; Fax: +1-202-994-287
| |
Collapse
|
44
|
Ma L, Li H, Zhuang H, Zhang Q, Peng N, Hu Y, Han N, Yang Y, Shi L. The Incidence of Metabolic Syndrome and the Valid Blood Pressure Cutoff Value for Predicting Metabolic Syndrome Within the Normal Blood Pressure Range in the Population Over 40 Years Old in Guiyang, China. Diabetes Metab Syndr Obes 2021; 14:2973-2983. [PMID: 34234494 PMCID: PMC8255645 DOI: 10.2147/dmso.s308923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/20/2021] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The study was conducted to investigate metabolic syndrome (MS) incidence within the normal blood pressure (BP) range in a population over 40 years old in an urban area of Guiyang, Guizhou Province, China, and to identify a valid BP cutoff value for predicting MS. MATERIALS AND METHODS Data from this study are based on the Risk Evaluation of cAncers in Chinese diabeTic Individuals: a lONgitudinal (REACTION) study. In total, 2935 subjects in the normal BP range and without MS (795 males, 2140 females) aged 40-80 years were included in this study. Follow-up subjects were classified by baseline BP and grouped by age. After the 3-year follow-up, the MS incidence and valid BP cutoff value for predicting MS were calculated for subjects within the normal BP range. RESULTS After the 3-year follow-up study, the crude and standardized MS incidences in the cohort were 19.83% and 16.89% for follow-up subjects, respectively, with incidences of 10.94% and 10.50% for males and 23.13% and 20.66% for females. The incidence of MS in normotensive subjects (15.53%) was lower than that in subjects with high-normal BP (29.08%). After adjusting for age, sex, BMI, smoking, and drinking, the risk of having MS in the high-normal BP group was 1.823-fold [HR 1.823 95% CI (1.538-2.162)] higher than that in the normotensive group. The ROC curve showed that the BP cutoff values were more than 117/74 mmHg in males and 112/74 mmHg in females. CONCLUSION When BP was within the normal range, the incidence of MS in participants was very high. The MS incidence was higher among high-normal BP subjects than among normotensive subjects, as was the risk of having MS. The valid BP cutoff value for predicting MS in the population was 117/74 mmHg in males and 112/74 mmHg in females. Currently, epidemiological investigations are needed to determine whether a lower BP cutoff value is needed in diagnosing MS.
Collapse
Affiliation(s)
- Li Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Hong Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- School Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Huijun Zhuang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Qiao Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Nianchun Peng
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Ying Hu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Na Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Yuxing Yang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Lixin Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| |
Collapse
|
45
|
Kirkman DL, Robinson AT, Rossman MJ, Seals DR, Edwards DG. Mitochondrial contributions to vascular endothelial dysfunction, arterial stiffness, and cardiovascular diseases. Am J Physiol Heart Circ Physiol 2021; 320:H2080-H2100. [PMID: 33834868 PMCID: PMC8163660 DOI: 10.1152/ajpheart.00917.2020] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/12/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) affects one in three adults and remains the leading cause of death in America. Advancing age is a major risk factor for CVD. Recent plateaus in CVD-related mortality rates in high-income countries after decades of decline highlight a critical need to identify novel therapeutic targets and strategies to mitigate and manage the risk of CVD development and progression. Vascular dysfunction, characterized by endothelial dysfunction and large elastic artery stiffening, is independently associated with an increased CVD risk and incidence and is therefore an attractive target for CVD prevention and management. Vascular mitochondria have emerged as an important player in maintaining vascular homeostasis. As such, age- and disease-related impairments in mitochondrial function contribute to vascular dysfunction and consequent increases in CVD risk. This review outlines the role of mitochondria in vascular function and discusses the ramifications of mitochondrial dysfunction on vascular health in the setting of age and disease. The adverse vascular consequences of increased mitochondrial-derived reactive oxygen species, impaired mitochondrial quality control, and defective mitochondrial calcium cycling are emphasized, in particular. Current evidence for both lifestyle and pharmaceutical mitochondrial-targeted strategies to improve vascular function is also presented.
Collapse
Affiliation(s)
- Danielle L Kirkman
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | | | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| |
Collapse
|
46
|
Levi DI, Wyrosdic JC, Hicks AI, Andrade MA, Toney GM, Prager-Khoutorsky M, Bourque CW. High dietary salt amplifies osmoresponsiveness in vasopressin-releasing neurons. Cell Rep 2021; 34:108866. [PMID: 33730577 PMCID: PMC8049100 DOI: 10.1016/j.celrep.2021.108866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/13/2020] [Accepted: 02/24/2021] [Indexed: 12/28/2022] Open
Abstract
High dietary salt increases arterial pressure partly through activation of magnocellular neurosecretory cells (MNCVP) that secrete the antidiuretic and vasoconstrictor hormone vasopressin (VP) into the circulation. Here, we show that the intrinsic and synaptic excitation of MNCVP caused by hypertonicity are differentially potentiated in two models of salt-dependent hypertension in rats. One model combined salty chow with a chronic subpressor dose of angiotensin II (AngII-salt), the other involved replacing drinking water with 2% NaCl (salt loading, SL). In both models, we observed a significant increase in the quantal amplitude of EPSCs on MNCVP. However, model-specific changes were also observed. AngII-salt increased the probability of glutamate release by osmoreceptor afferents and increased overall excitatory network drive. In contrast, SL specifically increased membrane stiffness and the intrinsic osmosensitivity of MNCVP. These results reveal that dietary salt increases the excitability of MNCVP through effects on the cell-autonomous and synaptic osmoresponsiveness of MNCVP.
Collapse
Affiliation(s)
- David I Levi
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G1A4, Canada
| | - Joshua C Wyrosdic
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G1A4, Canada
| | - Amirah-Iman Hicks
- Department of Physiology, McGill University, 3644 Promenade Sir William Osler, Montreal, QC H3G1Y6, Canada
| | - Mary Ann Andrade
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Centre San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Centre San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Masha Prager-Khoutorsky
- Department of Physiology, McGill University, 3644 Promenade Sir William Osler, Montreal, QC H3G1Y6, Canada.
| | - Charles W Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G1A4, Canada.
| |
Collapse
|
47
|
Souza LAC, Cooper SG, Worker CJ, Thakore P, Feng Earley Y. Use of chlorisondamine to assess the neurogenic contribution to blood pressure in mice: An evaluation of method. Physiol Rep 2021; 9:e14753. [PMID: 33587331 PMCID: PMC7883841 DOI: 10.14814/phy2.14753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 11/24/2022] Open
Abstract
Chlorisondamine (CSD) has been used to assess the neurogenic contribution to blood pressure (BP) and vasomotor sympathetic tone in animal models. It is assumed that the reduction in BP following CSD administration is associated to decreases in cardiac output (CO) and peripheral resistance, reflecting cardiac and vasomotor sympathetic tone, respectively. Surprisingly, this has not been characterized experimentally in mice, despite the extensive use of this animal model in cardiovascular research. We hypothesize that a specific dose of CSD can selectively block the sympathetic vasomotor tone. To test this hypothesis, we evaluated the effects of different doses of CSD (intraperitoneal) on BP and heart rate (HR) using telemetry, and on CO using echocardiography. BP and HR in normotensive C57Bl/6J mice reduced to a similar extent by all CSD doses tested (1-6 mg/kg). CSD at 6 mg/kg also reduced CO without affecting left ventricular stroke volume or fractional shortening. On the other hand, lower doses of CSD (1 and 2 mg/kg) produced significantly larger BP and HR reductions in DOCA-salt-induced hypertensive mice, indicating a greater neurogenic BP response. In addition, all doses of CSD reduced CO in hypertensive mice. Our data suggest that the BP response to CSD in mice likely reflects reduced CO and vasomotor sympathetic tone. We conclude that CSD can be used to assess the neurogenic contribution to BP in mice but may not be appropriate for specifically estimating vasomotor sympathetic tone.
Collapse
Affiliation(s)
- Lucas AC. Souza
- Departments of Pharmacology and Physiology & Cell BiologySchool of MedicineUniversity of Nevada, RenoRenoNVUSA
- Center for Molecular and Cellular Signaling in the Cardiovascular SystemUniversity of Nevada, RenoRenoNVUSA
| | - Silvana G. Cooper
- Departments of Pharmacology and Physiology & Cell BiologySchool of MedicineUniversity of Nevada, RenoRenoNVUSA
- Center for Molecular and Cellular Signaling in the Cardiovascular SystemUniversity of Nevada, RenoRenoNVUSA
| | - Caleb J. Worker
- Departments of Pharmacology and Physiology & Cell BiologySchool of MedicineUniversity of Nevada, RenoRenoNVUSA
- Center for Molecular and Cellular Signaling in the Cardiovascular SystemUniversity of Nevada, RenoRenoNVUSA
| | - Pratish Thakore
- Departments of Pharmacology and Physiology & Cell BiologySchool of MedicineUniversity of Nevada, RenoRenoNVUSA
- Center for Molecular and Cellular Signaling in the Cardiovascular SystemUniversity of Nevada, RenoRenoNVUSA
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell BiologySchool of MedicineUniversity of Nevada, RenoRenoNVUSA
- Center for Molecular and Cellular Signaling in the Cardiovascular SystemUniversity of Nevada, RenoRenoNVUSA
| |
Collapse
|
48
|
Kim DH, Kim KK, Lee TH, Eom H, Kim JW, Park JW, Jeong JK, Lee BJ. Transcription Factor TonEBP Stimulates Hyperosmolality-Dependent Arginine Vasopressin Gene Expression in the Mouse Hypothalamus. Front Endocrinol (Lausanne) 2021; 12:627343. [PMID: 33796071 PMCID: PMC8008816 DOI: 10.3389/fendo.2021.627343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/17/2021] [Indexed: 11/13/2022] Open
Abstract
The hypothalamic neuroendocrine system is strongly implicated in body energy homeostasis. In particular, the degree of production and release of arginine vasopressin (AVP) in the hypothalamus is affected by plasma osmolality, and that hypothalamic AVP is responsible for thirst and osmolality-dependent water and metabolic balance. However, the osmolality-responsive intracellular mechanism within AVP cells that regulates AVP synthesis is not clearly understood. Here, we report a role for tonicity-responsive enhancer binding protein (TonEBP), a transcription factor sensitive to cellular tonicity, in regulating osmosensitive hypothalamic AVP gene transcription. Our immunohistochemical work shows that hypothalamic AVP cellular activity, as recognized by c-fos, was enhanced in parallel with an elevation in TonEBP expression within AVP cells following water deprivation. Interestingly, our in vitro investigations found a synchronized pattern of TonEBP and AVP gene expression in response to osmotic stress. Those results indicate a positive correlation between hypothalamic TonEBP and AVP production during dehydration. Promoter and chromatin immunoprecipitation assays confirmed that TonEBP can bind directly to conserved binding motifs in the 5'-flanking promoter regions of the AVP gene. Furthermore, dehydration- and TonEBP-mediated hypothalamic AVP gene activation was reduced in TonEBP haploinsufficiency mice, compared with wild TonEBP homozygote animals. Therefore, our result support the idea that TonEBP is directly necessary, at least in part, for the elevation of AVP transcription in dehydration conditions. Additionally, dehydration-induced reductions in body weight were rescued in TonEBP haploinsufficiency mice. Altogether, our results demonstrate an intracellular machinery within hypothalamic AVP cells that is responsible for dehydration-induced AVP synthesis.
Collapse
Affiliation(s)
- Dong Hee Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Kwang Kon Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Tae Hwan Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Hyejin Eom
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Jin Kwon Jeong
- Department of Pharmacology and Physiology, School of Medicine & Health Sciences, The George Washington University, Washington, DC, United States
- *Correspondence: Byung Ju Lee, ; Jin Kwon Jeong,
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
- *Correspondence: Byung Ju Lee, ; Jin Kwon Jeong,
| |
Collapse
|
49
|
McKinley MJ, Pennington GL, Ryan PJ. The median preoptic nucleus: A major regulator of fluid, temperature, sleep, and cardiovascular homeostasis. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:435-454. [PMID: 34225980 DOI: 10.1016/b978-0-12-819975-6.00028-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Located in the midline lamina terminalis of the anterior wall of the third ventricle, the median preoptic nucleus is a thin elongated nucleus stretching around the rostral border of the anterior commissure. Its neuronal elements, composed of various types of excitatory glutamatergic and inhibitory GABAergic neurons, receive afferent neural signals from (1) neighboring subfornical organ and organum vasculosum of the lamina terminalis related to plasma osmolality and hormone concentrations, e.g., angiotensin II; (2) from peripheral sensors such as arterial baroreceptors and cutaneous thermosensors. Different sets of these MnPO glutamatergic and GABAergic neurons relay output signals to hypothalamic, midbrain, and medullary regions that drive homeostatic effector responses. Included in the effector responses are (1) thirst, antidiuretic hormone secretion and renal sodium excretion that subserve osmoregulation and body fluid homeostasis; (2) vasoconstriction or dilatation of skin blood vessels, and shivering and brown adipose tissue thermogenesis for core temperature homeostasis; (3) inhibition of hypothalamic and midbrain nuclei that stimulate wakefulness and arousal, thereby promoting both REM and non-REM sleep; and (4) activation of sympathetic pathways that drive vasoconstriction and heart rate to maintain arterial pressure and the perfusion of vital organs. The small size of MnPO belies its massive homeostatic significance.
Collapse
Affiliation(s)
- Michael J McKinley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| | - Glenn L Pennington
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Philip J Ryan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
50
|
Ribeiro N, Martins Sá RW, Antunes VR. Depletion of C1 neurons attenuates the salt-induced hypertension in unanesthetized rats. Brain Res 2020; 1748:147107. [PMID: 32905820 DOI: 10.1016/j.brainres.2020.147107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
High salt intake is able to evoke neuroendocrine and autonomic responses that include vasopressin release and sympathoexcitation resulting in increasing in the arterial blood pressure (BP). The C1 neurons are a specific population of catecholaminergic neurons located in the RVLM region and they control BP under homeostatic imbalance. Thus, here we hypothesized that the ablation of C1 neurons mitigate the high blood pressure induced by high-salt intake. To test this hypothesis, we injected anti-DβH-SAP saporin at the RVLM and monitored the BP in unanesthetized animals exposed to high salt intake of 2% NaCl solution for 7 days. The injection of anti-DβH-SAP into the RVLM depleted 80% of tyrosine hydroxylase-positive neurons (TH+ neurons) in the C1, 38% in the A5, and no significant reduction in the A1 region, when compared to control group (saline as vehicle). High salt intake elicited a significant increase in BP in the control group, while in the anti-DβH-SAP group the depletion of TH+ neurons prevents the salt-induced hypertension. Moreover, the low frequency component of systolic BP and pulse interval were increased by high-salt intake in control animals but not in anti-DβH-SAP group, which indirectly suggests that the increase in the BP is mediated by increase in sympathetic activity. In conclusion, our data show that hypertension induced by high-salt intake is dependent on C1 neurons.
Collapse
Affiliation(s)
- Natalia Ribeiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Renato W Martins Sá
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Vagner R Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|