1
|
Ventura RR, Ruginsk SG, Lopes da Silva A, Badauê-Passos D, Francescato HD, Coimbra TM, Elias LLK, Antunes-Rodrigues J. Interaction of glucocorticoids and interleukins in the control of hypothalamic neurohypophysial system output in salt loaded male rats. Neuropeptides 2025; 111:102523. [PMID: 40349477 DOI: 10.1016/j.npep.2025.102523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
The present study investigated the effects of a 4-day salt load (0.3 M NaCl, SL) and dexamethasone treatment (DEXA, 1 mg/Kg, subcutaneous) on the mechanisms possibly underlying glucocorticoid-mediated effects on hypothalamic neurohypophyseal system (HNS) activity. As expected, SL animals developed hyperosmolality, reflecting the progressive increase in plasma sodium concentrations. SL also triggered increased hypothalamic expression of vasopressin (AVP) and oxytocin (OT) messenger RNAs (mRNAs), increased magnocellular neuronal activation, and enhanced plasma hormone concentrations. Plasma corticosterone, interleukin (IL) 1β and tumor necrosis factor alfa, but not IL-6 levels, were also elevated in response to SL. Increased salt consumption also significantly decreased hypothalamic mRNA expression for the p65 subunit of the nuclear factor kappa B (NFkB), and increased mRNA expression for type β NFkB inhibitory protein (IκBβ). The protein expression ratio between phosphorylated and total NFκB was also elevated in SL rats. DEXA administration, in turn, prevented SL-induced AVP and OT release, as well as decreased corticosterone/IL plasma levels. Therefore, the present results suggest that increased salt consumption may originate a systemic-driven pro-inflammatory response, which can contribute to the increased secretion of corticosterone observed in SL animals. We therefore hypothesize that elevated systemic IL levels, in parallel with corticosterone secretion, may constitute, besides hyperosmolality, important redundant stimuli triggering SL-induced neuropeptide release. Conversely, high levels of corticosterone would produce, in the long term, inhibition of HNS activity and the termination of the neurosecretory response.
Collapse
Affiliation(s)
- R R Ventura
- Department of Physiological Sciences, Biomedical Sciences Institute, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil.
| | - S G Ruginsk
- Department of Physiological Sciences, Biomedical Sciences Institute, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - A Lopes da Silva
- Department of Physiology, Faculty of Medicine of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - D Badauê-Passos
- Laboratorio de Neuroendocrinologia Básica e Comportamental, Departamento de Fisiologia, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sergipe, São Cristovao, Sergipe, Brazil
| | - H D Francescato
- Department of Physiology, Faculty of Medicine of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - T M Coimbra
- Department of Physiology, Faculty of Medicine of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - L L K Elias
- Department of Physiology, Faculty of Medicine of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - J Antunes-Rodrigues
- Department of Physiology, Faculty of Medicine of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
2
|
Feng Y, Yin L, Li Y. BDNF-mediated depressor response by direct baroreceptor activation benefits for prevention and control of hypertension in high-latitude cold region. Neuropeptides 2025; 111:102506. [PMID: 40037144 DOI: 10.1016/j.npep.2025.102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Brian-derived neurotrophic factor (BDNF)-tyrosine kinase B (TrkB) signaling impacts on neuronal and cardiovascular physiology; however, its role in neurocontrol of circulation via baroreflex afferent pathway is largely unknown. Gene and protein expression of BDNF/TrkB were detected in the nodose (NG) and nucleus of tractus solitary (NTS) and expression levels were higher in male compared with female rats, which is relevant well with the blood pressure (BP, males > females in average). Microinjection of BDNF into NG dose-dependently reduced BP and this reduction was more dramatic in shamed control vs. renovascular hypertension (RVH) model rats, which partially inhibited in the presence of TrkB inhibitor K252a, indicating that BDNF-TrkB tends to lower BP under physiological and hypertensive conditions due presumably to a negative feed-back control by BP or compensatory mechanism. To answer this question, expression profiles for BDNF-TrkB were tested in the tissue of NG and NTS collected from RVH model rats. Consistently, the expression of both BDNF-TrkB were significantly up-regulated in RVH model alone with the elevation of BP. Taken these data together, our observation provides direct evidence showing the fundamental role of BDNF-TrkB signaling in autonomic control of BP regulation through baroreflex afferent function, potentially dominant role of BDNF-TrkB-mediated BP reduction in vivo baroreceptor activation due to distinct cellular mechanism compared with their role in the NTS, which extends our understanding of activity-dependent or compensatory mechanism of BDNF-TrkB in response to BP change, and sheds new light of BDNF-TrkB as potential target in prevention and control of hypertension in cold-region.
Collapse
Affiliation(s)
- Yan Feng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Lei Yin
- Department of Pharmacy, the 3(rd) Affiliated Hospital of Harbin medical University, Harbin, China
| | - Ying Li
- Department of Pharmacy, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China.
| |
Collapse
|
3
|
Zhang Z, He Z, Pan J, Yuan M, Lang Y, Wei X, Zhang C. The interaction of BDNF with estrogen in the development of hypertension and obesity, particularly during menopause. Front Endocrinol (Lausanne) 2024; 15:1384159. [PMID: 39655343 PMCID: PMC11625588 DOI: 10.3389/fendo.2024.1384159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
The expression of BDNF in both neuronal and non-neuronal cells is influenced by various stimuli, including prenatal developmental factors and postnatal conditions such as estrogens, dietary habits, and lifestyle factors like obesity, blood pressure, and aging. Central BDNF plays a crucial role in modulating how target tissues respond to these stimuli, influencing the pathogenesis of hypertension, mitigating obesity, and protecting neurons from aging. Thus, BDNF serves as a dynamic mediator of environmental influences, reflecting an individual's unique history of exposure. Estrogens, on the other hand, regulate various processes to maintain overall physiological well-being. Through nuclear estrogen receptors (ERα, ERβ) and the membrane estrogen receptor (GPER1), estrogens modulate transcriptional processes and signaling events that regulate the expression of target genes, such as ERα, components of the renin-angiotensin system (RAS), and hormone-sensitive lipase. Estrogens are instrumental in maintaining the set point for blood pressure and energy balance. BDNF and estrogens work cooperatively to prevent obesity by favoring lipolysis, and counteractively regulate blood pressure to adapt to the environment. Estrogen deficiency leads to menopause in women with low central BDNF level. This review delves into the complex mechanisms involving BDNF and estrogen, especially in the context of hypertension and obesity, particularly among postmenopausal women. The insights gained aim to inform the development of comprehensive therapeutic strategies for these prevalent syndromes affecting approximately 68% of adults.
Collapse
Affiliation(s)
- Zhongming Zhang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
- School of Medicine, Zhengzhou University of Industrial Technology, Xinzheng, Henan, China
| | - Ziyi He
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Jing Pan
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Minghui Yuan
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Yini Lang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Xiaomeng Wei
- School of Medicine, Zhengzhou University of Industrial Technology, Xinzheng, Henan, China
| | - Chaoyun Zhang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| |
Collapse
|
4
|
Joëls M, Karst H, Tasker JG. The emerging role of rapid corticosteroid actions on excitatory and inhibitory synaptic signaling in the brain. Front Neuroendocrinol 2024; 74:101146. [PMID: 39004314 DOI: 10.1016/j.yfrne.2024.101146] [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: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Over the past two decades, there has been increasing evidence for the importance of rapid-onset actions of corticosteroid hormones in the brain. Here, we highlight the distinct rapid corticosteroid actions that regulate excitatory and inhibitory synaptic transmission in the hypothalamus, the hippocampus, basolateral amygdala, and prefrontal cortex. The receptors that mediate rapid corticosteroid actions are located at or close to the plasma membrane, though many of the receptor characteristics remain unresolved. Rapid-onset corticosteroid effects play a role in fast neuroendocrine feedback as well as in higher brain functions, including increased aggression and anxiety, and impaired memory retrieval. The rapid non-genomic corticosteroid actions precede and complement slow-onset, long-lasting transcriptional actions of the steroids. Both rapid and slow corticosteroid actions appear to be indispensable to adapt to a continuously changing environment, and their imbalance can increase an individual's susceptibility to psychopathology.
Collapse
Affiliation(s)
- Marian Joëls
- University Medical Center Groningen, University of Groningen, the Netherlands; University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Henk Karst
- University Medical Center Utrecht, Utrecht University, the Netherlands; SILS-CNS. University of Amsterdam, the Netherlands.
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, and Southeast Louisiana Veterans Affairs Healthcare System, New Orleans, USA.
| |
Collapse
|
5
|
Kirchner MK, Althammer F, Donaldson KJ, Cox DN, Stern JE. Changes in neuropeptide large dense core vesicle trafficking dynamics contribute to adaptive responses to a systemic homeostatic challenge. iScience 2023; 26:108243. [PMID: 38026155 PMCID: PMC10654599 DOI: 10.1016/j.isci.2023.108243] [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: 05/31/2023] [Revised: 08/28/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Neuropeptides are packed into large dense core vesicles (LDCVs) that are transported from the soma out into their processes. Limited information exists regarding mechanisms regulating LDCV trafficking, particularly during challenges to bodily homeostasis. Addressing this gap, we used 2-photon imaging in an ex vivo preparation to study LDCVs trafficking dynamics in vasopressin (VP) neurons, which traffic and release neuropeptide from their dendrites and axons. We report a dynamic bidirectional trafficking of VP-LDCVs with important differences in speed and directionality between axons and dendrites. Acute, short-lasting stimuli known to alter VP firing activity and axonal/dendritic release caused modest changes in VP-LDCVs trafficking dynamics. Conversely, chronic/sustained systemic osmotic challenges upregulated VP-LDCVs trafficking dynamic, with a larger effect in dendrites. These results support differential regulation of dendritic and axonal LDCV trafficking, and that changes in trafficking dynamics constitute a novel mechanism by which peptidergic neurons can efficiently adapt to conditions of increased hormonal demand.
Collapse
Affiliation(s)
- Matthew K. Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30303, USA
- Institute of Human Genetics, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Kevin J. Donaldson
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Neuromics, Georgia State University, Atlanta, GA 30303, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Neuromics, Georgia State University, Atlanta, GA 30303, USA
| | - Javier E. Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
6
|
Nguyen DH, Duque V, Phillips N, Mecawi AS, Cunningham JT. Spatial transcriptomics reveal basal sex differences in supraoptic nucleus gene expression of adult rats related to cell signaling and ribosomal pathways. Biol Sex Differ 2023; 14:71. [PMID: 37858270 PMCID: PMC10585758 DOI: 10.1186/s13293-023-00554-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The supraoptic nucleus (SON) of the hypothalamus contains magnocellular neurosecretory cells that secrete the hormones vasopressin and oxytocin. Sex differences in SON gene expression have been relatively unexplored. Our study used spatially resolved transcriptomics to visualize gene expression profiles in the SON of adult male (n = 4) and female (n = 4) Sprague-Dawley rats using Visium Spatial Gene Expression (10x Genomics). METHODS Briefly, 10-μm coronal sections (~ 4 × 4 mm) containing the SON were collected from each rat and processed using Visium slides and recommended protocols. Data were analyzed using 10x Genomics' Space Ranger and Loupe Browser applications and other bioinformatic tools. Two unique differential expression (DE) analysis methods, Loupe Browser and DESeq2, were used. RESULTS Loupe Browser DE analysis of the SON identified 116 significant differentially expressed genes (DEGs) common to both sexes (e.g., Avp and Oxt), 31 significant DEGs unique to the males, and 73 significant DEGs unique to the females. DESeq2 analysis revealed 183 significant DEGs between the two groups. Gene Ontology (GO) enrichment and pathway analyses using significant genes identified via Loupe Browser revealed GO terms and pathways related to (1) neurohypophyseal hormone activity, regulation of peptide hormone secretion, and regulation of ion transport for the significant genes common to both males and females, (2) Gi signaling/G-protein mediated events for the significant genes unique to males, and (3) potassium ion transport/voltage-gated potassium channels for the significant genes unique to females, as some examples. GO/pathway analyses using significant genes identified via DESeq2 comparing female vs. male groups revealed GO terms/pathways related to ribosomal structure/function. Ingenuity Pathway Analysis (IPA) identified additional sex differences in canonical pathways (e.g., 'Mitochondrial Dysfunction', 'Oxidative Phosphorylation') and upstream regulators (e.g., CSF3, NFKB complex, TNF, GRIN3A). CONCLUSION There was little overlap in the IPA results for the two different DE methods. These results suggest sex differences in SON gene expression that are associated with cell signaling and ribosomal pathways.
Collapse
Affiliation(s)
- Dianna H Nguyen
- Department of Physiology and Anatomy, School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - Victor Duque
- Department of Biophysics, Laboratory of Molecular Neuroendocrinology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Nicole Phillips
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | - André Souza Mecawi
- Department of Biophysics, Laboratory of Molecular Neuroendocrinology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
7
|
Aikins AO, Farmer GE, Little JT, Cunningham JT. Effects of bile duct ligation on the inhibitory control of supraoptic vasopressin neurons. J Neuroendocrinol 2023; 35:e13312. [PMID: 37337093 PMCID: PMC10942741 DOI: 10.1111/jne.13312] [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: 02/17/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 06/21/2023]
Abstract
Dilutional hyponatremia due to increased plasma arginine vasopressin (AVP) is associated with liver cirrhosis. However, plasma AVP remains elevated despite progressive hypoosmolality. This study investigated changes to inhibitory control of supraoptic nucleus (SON) AVP neurons during liver cirrhosis. Experiments were conducted with adult male Sprague-Dawley rats. Bile duct ligation was used as a model of chronic liver cirrhosis. An adeno-associated virus containing a construct with an AVP promoter and either green fluorescent protein (GFP) or a ratiometric chloride indicator, ClopHensorN, was bilaterally injected into the SON of rats. After 2 weeks, rats received either BDL or sham surgery, and liver cirrhosis was allowed to develop for 4 weeks. In vitro, loose patch recordings of action potentials were obtained from GFP-labeled and unlabeled SON neurons in response to a brief focal application of the GABAA agonist muscimol (100 μM). Changes to intracellular chloride ([Cl]i) following muscimol application were determined by changes to the fluorescence ratio of ClopHensorN. The contribution of cation chloride cotransporters NKCC1 and KCC2 to changes in intracellular chloride was investigated using their respective antagonists, bumetanide (BU, 10 μM) and VU0240551 (10 μM). Plasma osmolality and hematocrit were measured as a marker of dilutional hyponatremia. The results showed reduced or absent GABAA -mediated inhibition in a greater proportion of AVP neurons from BDL rats as compared to sham rats (100% inhibition in sham vs. 47% in BDL, p = .001). Muscimol application was associated with increased [Cl]i in most cells from BDL as compared to cells from sham rats (χ2 = 30.24, p < .001). NKCC1 contributed to the impaired inhibition observed in BDL rats (p < .001 BDL - BU vs. BDL + BU). The results show that impaired inhibition of SON AVP neurons and increased intracellular chloride contribute to the sustained dilutional hyponatremia in liver cirrhosis.
Collapse
Affiliation(s)
- Ato O Aikins
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Joel T Little
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
8
|
Wu Q, Burley G, Li L, Lin S, Shi Y. The role of dietary salt in metabolism and energy balance: Insights beyond cardiovascular disease. Diabetes Obes Metab 2023; 25:1147-1161. [PMID: 36655379 PMCID: PMC10946535 DOI: 10.1111/dom.14980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
Dietary salt (NaCl) is essential to an organism's survival. However, today's diets are dominated by excessive salt intake, which significantly impacts individual and population health. High salt intake is closely linked to cardiovascular disease (CVD), especially hypertension, through a number of well-studied mechanisms. Emerging evidence indicates that salt overconsumption may also be associated with metabolic disorders. In this review, we first summarize recent updates on the mechanisms of salt-induced CVD, the effects of salt reduction and the use of salt substitution as a therapy. Next, we focus on how high salt intake can impact metabolism and energy balance, describing the mechanisms through which this occurs, including leptin resistance, the overproduction of fructose and ghrelin, insulin resistance and altered hormonal factors. A further influence on metabolism worth noting is the reported role of salt in inducing thermogenesis and increasing body temperature, leading to an increase in energy expenditure. While this result could be viewed as a positive metabolic effect because it promotes a negative energy balance to combat obesity, caution must be taken with this frame of thinking given the deleterious consequences of chronic high salt intake on cardiovascular health. Nevertheless, this review highlights the importance of salt as a noncaloric nutrient in regulating whole-body energy homeostasis. Through this review, we hope to provide a scientific framework for future studies to systematically address the metabolic impacts of dietary salt and salt replacement treatments. In addition, we hope to form a foundation for future clinical trials to explore how these salt-induced metabolic changes impact obesity development and progression, and to elucidate the regulatory mechanisms that drive these changes, with the aim of developing novel therapeutics for obesity and CVD.
Collapse
Affiliation(s)
- Qi Wu
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - George Burley
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Li‐Cheng Li
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Shu Lin
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Yan‐Chuan Shi
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
- School of Clinical Medicine, St Vincent's Clinical CampusFaculty of Medicine and HealthSydneyNew South WalesAustralia
| |
Collapse
|
9
|
Gomes PM, Batista JS, Sá RWM, Antunes VR. Short exposure to high salt in drinking solution leads to a cardiovascular phenotype of hypertension without changes in the blood volume of rats. Exp Physiol 2023; 108:361-370. [PMID: 36715005 PMCID: PMC10103861 DOI: 10.1113/ep090912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
NEW FINDINGS What is the central question of this study? Is the cardiovascular phenotype of high blood pressure observed in rats salt loaded with 2% NaCl in drinking solution a blood volume-dependent hypertension? What is the main finding and its importance? Animals exposed to 2% NaCl drinking solution develop hypertension, with dominance of sympathetic outflow and high [Na+ ] in the cerebrospinal fluid, but without changes in the blood volume. The phenotype of salt-dependent hypertension might be related to accumulation of [Na+ ] in the cerebrospinal fluid, which makes it an interesting animal model in which to study the neuronal pathways involved in control of the circulation in osmotic challenge conditions. ABSTRACT Evidence suggests that hypertension induced by high salt intake is correlated with an autonomic imbalance that favours sympathetic hyperactivity and an increase in vascular resistance, indicating a neurogenic component to this pathology. Although there are several animal models in which to study salt-induced hypertension with prolonged exposure to a high-sodium diet, here we sought to investigate whether the increase in arterial blood pressure of rats subjected to a short exposure to high salt, with 2% NaCl drinking solution instead of water, relies on changes in the circulating blood volume. Male Wistar rats were divided randomly into three groups: euhydrated (EU, n = 10), salt loaded (SL, n = 13) and water deprived (WD, n = 6). The SL rats exhibited a significant increase in mean arterial blood pressure, with a large low-frequency component of systolic arterial blood pressure variability, when compared with the EU group. Circulating blood volume did not differ between SL and EU rats, but it was lower in WD rats. Compared with EU rats, the [Na+ ] in cerebrospinal fluid was higher in SL rats and similar in magnitude to the WD rats. Plasma [Na+ ] did not differ between SL and EU rats, but it was higher in WD rats. Collectively, our data suggest that the hypertension induced by a short exposure to high salt intake closely resembles a neurogenic mechanism, but not a blood volume-dependent mechanism, with cumulative [Na+ ] in the cerebrospinal fluid that could be associated with changes in the neurochemistry of autonomic nuclei, which are highly susceptible to osmotic stress related to high salt consumption.
Collapse
Affiliation(s)
- Paula Magalhães Gomes
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| | - Julia Santos Batista
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| | - Renato Willian Martins Sá
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| | - Vagner Roberto Antunes
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| |
Collapse
|
10
|
Martin K, Toussaint ND, Tan SJ, Hewitson TD. Skin regulation of salt and blood pressure and potential clinical implications. Hypertens Res 2023; 46:408-416. [PMID: 36434290 DOI: 10.1038/s41440-022-01096-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Sodium chloride, as salt, gives rise to hypertension. Nevertheless, individual susceptibility to the ramifications of sodium chloride is heterogeneous. The conventional nephron-centric regulation of sodium with neurohormonal inputs and responses is now expanded to include an intricate extrarenal pathway including the endothelium, skin, lymphatics, and immune cells. An overabundance of sodium is buffered and regulated by the skin interstitium. Excess sodium passes through (and damages) the vascular endothelium and can be dynamically stored in the skin, modulated by skin immune cells and lymphatics. This excess interstitially stored sodium is implicated in hypertension, cardiovascular dysfunction, metabolic disruption, and inflammatory dysregulation. This extrarenal pathway of regulating sodium represents a novel target for better blood pressure management, rebalancing disturbed inflammation, and hence addressing cardiovascular and metabolic disease.
Collapse
Affiliation(s)
- Kylie Martin
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia. .,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.
| | - Nigel D Toussaint
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Sven-Jean Tan
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Timothy D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
11
|
Aikins AO, Little JT, Rybalchenko N, Cunningham JT. Norepinephrine innervation of the supraoptic nucleus contributes to increased copeptin and dilutional hyponatremia in male rats. Am J Physiol Regul Integr Comp Physiol 2022; 323:R797-R809. [PMID: 36189988 PMCID: PMC9639772 DOI: 10.1152/ajpregu.00086.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022]
Abstract
Dilutional hyponatremia associated with liver cirrhosis is due to inappropriate release of arginine vasopressin (AVP). Elevated plasma AVP causes water retention resulting in a decrease in plasma osmolality. Cirrhosis, in this study caused by ligation of the common bile duct (BDL), leads to a decrease in central vascular blood volume and hypotension, stimuli for nonosmotic AVP release. The A1/A2 neurons stimulate the release of AVP from the supraoptic nucleus (SON) in response to nonosmotic stimuli. We hypothesize that the A1/A2 noradrenergic neurons support chronic release of AVP in cirrhosis leading to dilutional hyponatremia. Adult, male rats were anesthetized with 2-3% isoflurane (mixed with 95% O2/5% CO2) and injected in the SON with anti-dopamine β-hydroxylase (DBH) saporin (DSAP) or vehicle followed by either BDL or sham surgery. Plasma copeptin, osmolality, and hematocrit were measured. Brains were processed for ΔFosB, dopamine β-hydroxylase (DBH), and AVP immunohistochemistry. DSAP injection: 1) significantly reduced the number of DBH immunoreactive A1/A2 neurons (A1, P < 0.0001; A2, P = 0.0014), 2) significantly reduced the number of A1/A2 neurons immunoreactive to both DBH and ΔFosB positive neurons (A1, P = 0.0015; A2, P < 0.0001), 3) reduced the number of SON neurons immunoreactive to both AVP and ΔFosB (P < 0.0001), 4) prevented the increase in plasma copeptin observed in vehicle-injected BDL rats (P = 0.0011), and 5) normalized plasma osmolality and hematocrit (plasma osmolality, P = 0.0475; hematocrit, P = 0.0051) as compared with vehicle injection. Our data suggest that A1/A2 neurons contribute to increased plasma copeptin and hypoosmolality in male BDL rats.
Collapse
Affiliation(s)
- Ato O Aikins
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - Joel T Little
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - Nataliya Rybalchenko
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| |
Collapse
|
12
|
Kurtz T, Pravenec M, DiCarlo S. Mechanism-based strategies to prevent salt sensitivity and salt-induced hypertension. Clin Sci (Lond) 2022; 136:599-620. [PMID: 35452099 PMCID: PMC9069470 DOI: 10.1042/cs20210566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
High-salt diets are a major cause of hypertension and cardiovascular (CV) disease. Many governments are interested in using food salt reduction programs to reduce the risk for salt-induced increases in blood pressure and CV events. It is assumed that reducing the salt concentration of processed foods will substantially reduce mean salt intake in the general population. However, contrary to expectations, reducing the sodium density of nearly all foods consumed in England by 21% had little or no effect on salt intake in the general population. This may be due to the fact that in England, as in other countries including the U.S.A., mean salt intake is already close to the lower normal physiologic limit for mean salt intake of free-living populations. Thus, mechanism-based strategies for preventing salt-induced increases in blood pressure that do not solely depend on reducing salt intake merit attention. It is now recognized that the initiation of salt-induced increases in blood pressure often involves a combination of normal increases in sodium balance, blood volume and cardiac output together with abnormal vascular resistance responses to increased salt intake. Therefore, preventing either the normal increases in sodium balance and cardiac output, or the abnormal vascular resistance responses to salt, can prevent salt-induced increases in blood pressure. Suboptimal nutrient intake is a common cause of the hemodynamic disturbances mediating salt-induced hypertension. Accordingly, efforts to identify and correct the nutrient deficiencies that promote salt sensitivity hold promise for decreasing population risk of salt-induced hypertension without requiring reductions in salt intake.
Collapse
Affiliation(s)
- Theodore W. Kurtz
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94017-0134, U.S.A
| | - Michal Pravenec
- Institute of Physiology, Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Stephen E. DiCarlo
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, U.S.A
| |
Collapse
|
13
|
Rauf S, Achmad I, Lestaluhu SA. WATER SUPPLEMENTATION AFFECTS THE FLUID INTAKE AND SERUM LEVELS OF BDNF AMONG THE ELDERLY IN NURSING HOME. Nutrition 2022; 101:111700. [DOI: 10.1016/j.nut.2022.111700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/29/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
|
14
|
Zhang C, Lin Y, Wu Q, Yan C, Wong MW, Zeng F, Zhu P, Bowes K, Lee K, Zhang X, Song Z, Lin S, Shi Y. Arcuate NPY is involved in salt‐induced hypertension via modulation of paraventricular vasopressin and brain‐derived neurotrophic factor. J Cell Physiol 2022; 237:2574-2588. [PMID: 35312067 PMCID: PMC9544553 DOI: 10.1002/jcp.30719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 12/17/2022]
Abstract
Chronic high salt intake is one of the leading causes of hypertension. Salt activates the release of the key neurotransmitters in the hypothalamus such as vasopressin to increase blood pressure, and neuropepetide Y (NPY) has been implicated in the modulation of vasopressin levels. NPY in the hypothalamic arcuate nucleus (Arc) is best known for its control in appetite and energy homeostasis, but it is unclear whether it is also involved in the development of salt‐induced hypertension. Here, we demonstrate that wild‐type mice given 2% NaCl salt water for 8 weeks developed hypertension which was associated with marked downregulation of NPY expression in the hypothalamic Arc as demonstrated in NPY‐GFP reporter mice as well as by in situ hybridization analysis. Furthermore, salt intake activates neurons in the hypothalamic paraventricular nucleus (PVN) where mRNA expression of brain‐derived neurotrophic factor (BDNF) and vasopressin was found to be upregulated, leading to elevated serum vasopressin levels. This finding suggests an inverse correlation between the Arc NPY level and expression of vasopressin and BDNF in the PVN. Specific restoration of NPY by injecting AAV‐Cre recombinase into the Arc only of the NPY‐targeted mutant mice carrying a loxP‐flanked STOP cassette reversed effects of salt intake on vasopressin and BDNF expression, leading to a normalization of salt‐dependent blood pressure. In summary, our study uncovers an important Arc NPY‐originated neuronal circuitry that could sense and respond to peripheral electrolyte signals and thereby regulate hypertension via vasopressin and BDNF in the PVN.
Collapse
Affiliation(s)
- Chen‐Liang Zhang
- Department of Cardiology, Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Yi‐Zhang Lin
- Department of Cardiology, Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Qi Wu
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
- The Second Affiliated Hospital Fujian Medical University Quanzhou China
| | - Chenxu Yan
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
- The Second Affiliated Hospital Fujian Medical University Quanzhou China
| | - Matthew Wai‐Kin Wong
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
| | - Fan Zeng
- Department of Cardiology, Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Ping Zhu
- Department of Cardiology, Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Kelsey Bowes
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
| | - Kailun Lee
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
| | - Xuan Zhang
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
| | - Zhi‐Yuan Song
- Department of Cardiology, Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Shu Lin
- Department of Cardiology, Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
- The Second Affiliated Hospital Fujian Medical University Quanzhou China
| | - Yan‐Chuan Shi
- Group of Neuroendocrinology, Diabetes and Metabolism Division Garvan Institute of Medical Research Sydney New South Wales Australia
- St Vincent's Clinical School UNSW Sydney Sydney New South Wales Australia
| |
Collapse
|
15
|
Wu H, Sun Q, Yuan S, Wang J, Li F, Gao H, Chen X, Yang R, Xu J. AT1 Receptors: Their Actions from Hypertension to Cognitive Impairment. Cardiovasc Toxicol 2022; 22:311-325. [PMID: 35211833 PMCID: PMC8868040 DOI: 10.1007/s12012-022-09730-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022]
Abstract
Hypertension is one of the most prevalent cardiovascular disorders worldwide, affecting 1.13 billion people, or 14% of the global population. Hypertension is the single biggest risk factor for cerebrovascular dysfunction. According to the American Heart Association, high blood pressure (BP), especially in middle-aged individuals (~ 40 to 60 years old), is associated with an increased risk of dementia, later in life. Alzheimer’s disease and cerebrovascular disease are the two leading causes of dementia, accounting for around 80% of the total cases and usually combining mixed pathologies from both. Little is known regarding how hypertension affects cognitive function, so the impact of its treatment on cognitive impairment has been difficult to assess. The brain renin-angiotensin system (RAS) is essential for BP regulation and overactivity of this system has been established to precede the development and maintenance of hypertension. Angiotensin II (Ang-II), the main peptide within this system, induces vasoconstriction and impairs neuro-vascular coupling by acting on brain Ang-II type 1 receptors (AT1R). In this review, we systemically analyzed the association between RAS and biological mechanisms of cognitive impairment, from the perspective of AT1R located in the central nervous system. Additionally, the possible contribution of brain AT1R to global cognition decline in COVID-19 cases will be discussed as well.
Collapse
Affiliation(s)
- Hanxue Wu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Qi Sun
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shenglan Yuan
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Jiawei Wang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Fanni Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongli Gao
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Rui Yang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Jiaxi Xu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
16
|
Jiang S, Wang YQ, Tang Y, Lu X, Guo D. Environmental Enrichment Protects Against Sepsis-Associated Encephalopathy-Induced Learning and Memory Deficits by Enhancing the Synthesis and Release of Vasopressin in the Supraoptic Nucleus. J Inflamm Res 2022; 15:363-379. [PMID: 35079222 PMCID: PMC8776728 DOI: 10.2147/jir.s345108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
Background As a severe complication of sepsis, sepsis-associated encephalopathy (SAE) usually manifests as impaired learning and memory ability in survivors. Previous studies have reported that environmental enrichment (EE) can increase the learning and memory ability in different brain injury models. However, there has been no research on the possible positive effect of EE on SAE. Aim The present study aimed to test the effect of EE on SAE-induced impairment of learning and memory and its related mechanisms. Methods A Morris water maze test (MWM) was used to evaluate the learning and memory ability of SAE rats that received EE housing or not. The expression of vasopressin (VP) was assessed using immunofluorescence microscopy and enzyme-linked immunosorbent assays (ELISAs). The synthesis of VP in the supraoptic nucleus (SON) was determined using quantitative real-time reverse transcription-PCR analysis. Moreover, inflammatory markers and brain-derived neurotrophic factor (BDNF) were detected using ELISA. Results The results showed that SAE induced a decreased learning and memory ability, while EE reversed this impairment. EE also enhanced the synthesis and secretion of VP in the SON. Blocking the action of VP in the hippocampus interrupted the EE-induced amelioration of learning and memory impairment. Moreover, EE induced changes to the levels of BDNF and cytokines in the hippocampus and these effects were mediated by VP binding to the VP receptor 1a. Conclusion Our findings demonstrated that the enhanced synthesis and secretion of VP in the SON are a key determinant responsible for EE-induced alleviation of learning and memory deficits caused by SAE.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Rehabilitation Medicine, the China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
- Correspondence: Shan Jiang, Department of Rehabilitation Medicine, the China-Japan Friendship Hospital, No. 2 Ying Hua Yuan East Street, Beijing, 100029, People’s Republic of China, Tel +86 10 84205288, Fax +86 10 64217749, Email
| | - Yong-Qiang Wang
- Department of Ophthalmology, the Sunshine Union Hospital, Weifang, Shandong, 261071, People’s Republic of China
| | - Yifei Tang
- Department of Rehabilitation Medicine, the China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Xi Lu
- Department of Rehabilitation Medicine, the China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Dan Guo
- Department of Rehabilitation Medicine, the China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| |
Collapse
|
17
|
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
|
18
|
Thorsdottir D, Einwag Z, Erdos B. BDNF shifts excitatory-inhibitory balance in the paraventricular nucleus of the hypothalamus to elevate blood pressure. J Neurophysiol 2021; 126:1209-1220. [PMID: 34406887 DOI: 10.1152/jn.00247.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Presympathetic neurons in the paraventricular nucleus of the hypothalamus (PVN) play a key role in cardiovascular regulation. We have previously shown that brain-derived neurotrophic factor (BDNF), acting in the PVN, increases sympathetic activity and blood pressure and serves as a key regulator of stress-induced hypertensive responses. BDNF is known to alter glutamatergic and GABA-ergic signaling broadly in the central nervous system, but whether BDNF has similar actions in the PVN remains to be investigated. Here, we tested the hypothesis that increased BDNF expression in the PVN elevates blood pressure by enhancing N-methyl-d-aspartate (NMDA) receptor (NMDAR)- and inhibiting GABAA receptor (GABAAR)-mediated signaling. Sprague-Dawley rats received bilateral PVN injections of AAV2 viral vectors expressing green fluorescent protein (GFP) or BDNF. Three weeks later, cardiovascular responses to PVN injections of NMDAR and GABAAR agonists and antagonists were recorded under α-chloralose-urethane anesthesia. In addition, expressions of excitatory and inhibitory signaling components in the PVN were assessed using immunofluorescence. Our results showed that NMDAR inhibition led to a greater decrease in blood pressure in the BDNF vs. GFP group, while GABAAR inhibition led to greater increases in blood pressure in the GFP group compared to BDNF. Conversely, GABAAR activation decreased blood pressure significantly more in GFP vs. BDNF rats. In addition, immunoreactivity of NMDAR1 was upregulated, while GABAAR-α1 and K+/Cl- cotransporter 2 were downregulated by BDNF overexpression in the PVN. In summary, our findings indicate that hypertensive actions of BDNF within the PVN are mediated, at least in part, by augmented NMDAR and reduced GABAAR signaling.NEW & NOTEWORTHY We have shown that BDNF, acting in the PVN, elevates blood pressure in part by augmenting NMDA receptor-mediated excitatory input and by diminishing GABAA receptor-mediated inhibitory input to PVN neurons. In addition, we demonstrate that elevated BDNF expression in the PVN upregulates NMDA receptor immunoreactivity and downregulates GABAA receptor as well as KCC2 transporter immunoreactivity.
Collapse
Affiliation(s)
| | - Zachary Einwag
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| |
Collapse
|
19
|
Dinh QN, Drummond GR, Sobey CG. Estrogen: reducing the pressure by arginine vasopressin. Cardiovasc Res 2021; 117:2143-2144. [PMID: 33351906 DOI: 10.1093/cvr/cvaa341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Quynh Nhu Dinh
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
20
|
Jin X, Kim WB, Kim MN, Jung WW, Kang HK, Hong EH, Kim YS, Shim WJ, Han HC, Colwell CS, Kim YB, Kim YI. Oestrogen inhibits salt-dependent hypertension by suppressing GABAergic excitation in magnocellular AVP neurons. Cardiovasc Res 2021; 117:2263-2274. [PMID: 32960965 PMCID: PMC10616626 DOI: 10.1093/cvr/cvaa271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/24/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Abundant evidence indicates that oestrogen (E2) plays a protective role against hypertension. Yet, the mechanism underlying the antihypertensive effect of E2 is poorly understood. In this study, we sought to determine the mechanism through which E2 inhibits salt-dependent hypertension. METHODS AND RESULTS To this end, we performed a series of in vivo and in vitro experiments employing a rat model of hypertension that is produced by deoxycorticosterone acetate (DOCA)-salt treatment after uninephrectomy. We found that E2 prevented DOCA-salt treatment from inducing hypertension, raising plasma arginine-vasopressin (AVP) level, enhancing the depressor effect of the V1a receptor antagonist (Phenylac1,D-Tyr(Et)2,Lys6,Arg8,des-Gly9)-vasopressin, and converting GABAergic inhibition to excitation in hypothalamic magnocellular AVP neurons. Moreover, we obtained results indicating that the E2 modulation of the activity and/or expression of NKCC1 (Cl- importer) and KCC2 (Cl- extruder) underpins the effect of E2 on the transition of GABAergic transmission in AVP neurons. Lastly, we discovered that, in DOCA-salt-treated hypertensive ovariectomized rats, CLP290 (prodrug of the KCC2 activator CLP257, intraperitoneal injections) lowered blood pressure, and plasma AVP level and hyperpolarized GABA equilibrium potential to prevent GABAergic excitation from emerging in the AVP neurons of these animals. CONCLUSION Based on these results, we conclude that E2 inhibits salt-dependent hypertension by suppressing GABAergic excitation to decrease the hormonal output of AVP neurons.
Collapse
Affiliation(s)
- Xiangyan Jin
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Woong Bin Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Mi-Na Kim
- Department of Internal Medicine, Cardiovascular Section, Korea University Anam Hospital, Seoul 136-705, Republic of Korea
| | - Won Woo Jung
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Hyung Kyung Kang
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Eun-Hwa Hong
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Yoon Sik Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Wan Joo Shim
- Department of Internal Medicine, Cardiovascular Section, Korea University Anam Hospital, Seoul 136-705, Republic of Korea
| | - Hee Chul Han
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Young-Beom Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Yang In Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| |
Collapse
|
21
|
Aikins AO, Nguyen DH, Paundralingga O, Farmer GE, Shimoura CG, Brock C, Cunningham JT. Cardiovascular Neuroendocrinology: Emerging Role for Neurohypophyseal Hormones in Pathophysiology. Endocrinology 2021; 162:6247962. [PMID: 33891015 PMCID: PMC8234498 DOI: 10.1210/endocr/bqab082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 11/19/2022]
Abstract
Arginine vasopressin (AVP) and oxytocin (OXY) are released by magnocellular neurosecretory cells that project to the posterior pituitary. While AVP and OXY currently receive more attention for their contributions to affiliative behavior, this mini-review discusses their roles in cardiovascular function broadly defined to include indirect effects that influence cardiovascular function. The traditional view is that neither AVP nor OXY contributes to basal cardiovascular function, although some recent studies suggest that this position might be re-evaluated. More evidence indicates that adaptations and neuroplasticity of AVP and OXY neurons contribute to cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Ato O Aikins
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Dianna H Nguyen
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Obed Paundralingga
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - George E Farmer
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Caroline Gusson Shimoura
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Courtney Brock
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: J. Thomas Cunningham Department of Physiology & Anatomy CBH 338 UNT Health Science Center 3500 Camp Bowie Blvd Fort Worth, TX 76107, USA.
| |
Collapse
|
22
|
Yang X, Liu S, Zhang Z. Sex difference in blood pressure, a combinatorial consequence of the differential in RAAS components, sex hormones and time course. Curr Hypertens Rev 2021; 18:11-16. [PMID: 33992057 DOI: 10.2174/1573402117666210511011444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/26/2021] [Accepted: 03/02/2021] [Indexed: 11/22/2022]
Abstract
The longitudinal increment of blood pressure (BP) with age is attributed to lifestyle, internal and external environments. It is not limited to systemic brain-derived neurotrophic factor (BDNF), signaling to allow the individuals to better adapt to the developmental and environmental change. This regulation is necessary for all lives, regardless of sex. Basic levels of renin-angiotensin- aldosterone system (RAAS) components in males and females define the fundamental sex difference in BP, which may be set by prenatal programming and profoundly influence BP after birth. The innate sex difference in BP is magnified during puberty growth and further modified by menopause. At the age of 70 or older, blood pressure was similar in men and women. The understanding of the prenatal setup and development of sexual dimorphism in BP may provide preventative therapeutic strategies, including timing and drugs, for individuals with abnormal BP.
Collapse
Affiliation(s)
- Xiaomei Yang
- College of Basic Medicine, Henan University of Chinese Medicine,156,Jinshui East Road ,Zhengdong New District, Zhengzhou, Henan 450046. China
| | - Shien Liu
- College of Basic Medicine, Henan University of Chinese Medicine,156,Jinshui East Road ,Zhengdong New District, Zhengzhou, Henan 450046. China
| | - Zhongming Zhang
- College of Chinese Medicine, and Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology; 80 Changjiang Rd. Nanyang, Henan, 473004. China
| |
Collapse
|
23
|
Marciante AB, Shell B, Farmer GE, Cunningham JT. Role of angiotensin II in chronic intermittent hypoxia-induced hypertension and cognitive decline. Am J Physiol Regul Integr Comp Physiol 2021; 320:R519-R525. [PMID: 33595364 PMCID: PMC8238144 DOI: 10.1152/ajpregu.00222.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 01/19/2021] [Accepted: 02/13/2021] [Indexed: 02/03/2023]
Abstract
Sleep apnea is characterized by momentary interruptions in normal respiration and leads to periods of decreased oxygen, or intermittent hypoxia. Chronic intermittent hypoxia is a model of the hypoxemia associated with sleep apnea and results in a sustained hypertension that is maintained during normoxia. Adaptations of the carotid body and activation of the renin-angiotensin system may contribute to the development of hypertension associated with chronic intermittent hypoxia. The subsequent activation of the brain renin-angiotensin system may produce changes in sympathetic regulatory neural networks that support the maintenance of the hypertension associated with intermittent hypoxia. Hypertension and sleep apnea not only increase risk for cardiovascular disease but are also risk factors for cognitive decline and Alzheimer's disease. Activation of the angiotensin system could be a common mechanism that links these disorders.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing REsearch And THErapeutics (BREATHE) Center, University of Florida, Gainesville, Florida
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Brent Shell
- Zuckerberg College of Health Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
24
|
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
|
25
|
Hasegawa Y, Takemoto Y, Hayashi K, Kameno K, Kim-Mitsuyama S. The endogenous and exogenous brain-derived neurotrophic factor plays pivotal roles in the pathogenesis of stroke onset in high salt-loaded hypertensive rats. Exp Gerontol 2021; 147:111286. [PMID: 33609688 DOI: 10.1016/j.exger.2021.111286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/23/2021] [Accepted: 02/14/2021] [Indexed: 12/17/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is known to have neuroprotective effects on multiple neurovascular diseases especially poststroke recovery. On the other hand, BDNF reported to increase blood pressure (BP) which is one of the major risk factors for stroke onset. To clarify the conflicting effects on stroke onset, we examined the expression of endogenous BDNF in relation to stroke onset. In addition, we explored the effect of exogenous central BDNF against stroke onset and all-cause mortality as the primary endpoint and BP as the secondary object in hypertensive rats with high-salt diet. In experiment 1, male spontaneously hypertensive stroke-prone rats (SHRSP) were fed a 0.3% (n = 8) or an 8% (n = 22) sodium diet (Na) through 28 days. The SHRSP with 8% Na showed significant increase of stroke onset, all-cause mortality, upregulation of reactive astrocytes, and disruption of blood-brain barrier. BDNF in the rats with 8% Na was significantly upregulated and mainly expressed in reactive astrocytes, whereas phosphorylated tropomyosin-related kinase B did not change by the rich BDNF. In experiment 2, male SHRSP were treated with continuous intracerebroventricular injection of 2.1 μg/day BDNF (n = 10) or the vehicle (Phosphate buffer saline; n = 10) and fed an 8% Na through 24 days. Exogenous central BDNF induced significant increase of BP and heart rate, and exhibited higher stroke onset and all-cause mortality compared with vehicle group. The present study demonstrated that endogenous BDNF were significantly produced in reactive astrocytes in relation to stroke onset regardless of neuroprotection. In addition, exogenous central BDNF increased BP which might be associated with sympathetic nerve activity and provided unfavorable effects on the prognosis of hypertensive rats. As BDNF is still potentially a good candidate for the treatment of neurovascular diseases, we suggest that hypertensive patients need care for the elevation of BP in the clinical trials of BDNF.
Collapse
Affiliation(s)
- Yu Hasegawa
- Departments of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Kumamoto, Kumamoto, Japan; Department of Pharmaceutical Science, School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1, Enokizu, Okawa, Fukuoka 8318501, Japan.
| | - Yushin Takemoto
- Departments of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Kumamoto, Kumamoto, Japan
| | - Kenyu Hayashi
- Departments of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Kumamoto, Kumamoto, Japan
| | - Koki Kameno
- Departments of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Kumamoto, Kumamoto, Japan
| | - Shokei Kim-Mitsuyama
- Departments of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Kumamoto, Kumamoto, Japan
| |
Collapse
|
26
|
Bordeleau M, Fernández de Cossío L, Chakravarty MM, Tremblay MÈ. From Maternal Diet to Neurodevelopmental Disorders: A Story of Neuroinflammation. Front Cell Neurosci 2021; 14:612705. [PMID: 33536875 PMCID: PMC7849357 DOI: 10.3389/fncel.2020.612705] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Providing the appropriate quantity and quality of food needed for both the mother's well-being and the healthy development of the offspring is crucial during pregnancy. However, the macro- and micronutrient intake also impacts the body's regulatory supersystems of the mother, such as the immune, endocrine, and nervous systems, which ultimately influence the overall development of the offspring. Of particular importance is the association between unhealthy maternal diet and neurodevelopmental disorders in the offspring. Epidemiological studies have linked neurodevelopmental disorders like autism spectrum disorders, attention-deficit-hyperactivity disorder, and schizophrenia, to maternal immune activation (MIA) during gestation. While the deleterious consequences of diet-induced MIA on offspring neurodevelopment are increasingly revealed, neuroinflammation is emerging as a key underlying mechanism. In this review, we compile the evidence available on how the mother and offspring are both impacted by maternal dietary imbalance. We specifically explore the various inflammatory and anti-inflammatory effects of dietary components and discuss how changes in inflammatory status can prime the offspring brain development toward neurodevelopmental disorders. Lastly, we discuss research evidence on the mechanisms that sustain the relationship between maternal dietary imbalance and offspring brain development, involving altered neuroinflammatory status in the offspring, as well as genetic to cellular programming notably of microglia, and the evidence that the gut microbiome may act as a key mediator.
Collapse
Affiliation(s)
- Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | - M. Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Cerebral Imaging Centre, Douglas Mental Health University, McGill University, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Wen S, Nguyen T, Gong M, Yuan X, Wang C, Jin J, Zhou L. An Overview of Similarities and Differences in Metabolic Actions and Effects of Central Nervous System Between Glucagon-Like Peptide-1 Receptor Agonists (GLP-1RAs) and Sodium Glucose Co-Transporter-2 Inhibitors (SGLT-2is). Diabetes Metab Syndr Obes 2021; 14:2955-2972. [PMID: 34234493 PMCID: PMC8254548 DOI: 10.2147/dmso.s312527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
GLP-1 receptor agonists (GLP-1RAs) and SGLT-2 inhibitors (SGLT-2is) are novel antidiabetic medications associated with considerable cardiovascular benefits therapying treatment of diabetic patients. GLP-1 exhibits atherosclerosis resistance, whereas SGLT-2i acts to ameliorate the neuroendocrine state in the patients with chronic heart failure. Despite their distinct modes of action, both factors share pathways by regulating the central nervous system (CNS). While numerous preclinical and clinical studies have demonstrated that GLP-1 can access various nuclei associated with energy homeostasis and hedonic eating in the CNS via blood-brain barrier (BBB), research on the activity of SGLT-2is remains limited. In our previous studies, we demonstrated that both GLP-1 receptor agonists (GLP-1RAs) liraglutide and exenatide, as well as an SGLT-2i, dapagliflozin, could activate various nuclei and pathways in the CNS of Sprague Dawley (SD) rats and C57BL/6 mice, respectively. Moreover, our results revealed similarities and differences in neural pathways, which possibly regulated different metabolic effects of GLP-1RA and SGLT-2i via sympathetic and parasympathetic systems in the CNS, such as feeding, blood glucose regulation and cardiovascular activities (arterial blood pressure and heart rate control). In the present article, we extensively discuss recent preclinical studies on the effects of GLP-1RAs and SGLT-2is on the CNS actions, with the aim of providing a theoretical explanation on their mechanism of action in improvement of the macro-cardiovascular risk and reducing incidence of diabetic complications. Overall, these findings are expected to guide future drug design approaches.
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Thiquynhnga Nguyen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Jianlan Jin
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of ChinaTel +8613611927616 Email
| |
Collapse
|
28
|
Kim YB, Jung WW, Lee SW, Jin X, Kang HK, Hong EH, Min SS, Kim YS, Han HC, Colwell CS, Kim YI. Excessive maternal salt intake gives rise to vasopressin-dependent salt sensitivity of blood pressure in male offspring. J Mol Cell Cardiol 2021; 150:12-22. [PMID: 33011158 DOI: 10.1016/j.yjmcc.2020.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/28/2020] [Accepted: 09/26/2020] [Indexed: 01/11/2023]
Abstract
Salt sensitivity of blood pressure (SSBP) is a trait carrying strong prognostic implications for various cardiovascular diseases. To test the hypothesis that excessive maternal salt intake causes SSBP in offspring through a mechanism dependent upon arginine-vasopressin (AVP), we performed a series of experiments using offspring of the rat dams salt-loaded during pregnancy and lactation with 1.5% saline drink ("experimental offspring") and those with normal perinatal salt exposure ("control offspring"). Salt challenge, given at 7-8 weeks of age with either 2% saline drink (3 days) or 8% NaCl-containing chow (4 weeks), had little or no effect on systolic blood pressure (SBP) in female offspring, whereas the salt challenge significantly raised SBP in male offspring, with the magnitude of increase being greater in experimental, than control, rats. Furthermore, the salt challenge not only raised plasma AVP level more and caused greater depressor responses to V1a and V2 AVP receptor antagonists to occur in experimental, than control, males, but it also made GABA excitatory in a significant proportion of magnocellular AVP neurons of experimental males by depolarizing GABA equilibrium potential. The effect of the maternal salt loading on the salt challenge-elicited SBP response in male offspring was precluded by maternal conivaptan treatment (non-selective AVP receptor antagonist) during the salt-loading period, whereas it was mimicked by neonatal AVP treatment. These results suggest that the excessive maternal salt intake brings about SSBP in male offspring, both the programming and the expression of which depend on increased AVP secretion that may partly result from excitatory GABAergic action.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Won Woo Jung
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Won Lee
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Xiangyan Jin
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyung Kyung Kang
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Eun-Hwa Hong
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sun Seek Min
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon 34824, Republic of Korea
| | - Yoon-Sik Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hee Chul Han
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California School of Medicine, Los Angeles, CA, United States of America.
| | - Yang In Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| |
Collapse
|
29
|
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
|
30
|
Meor Azlan NF, Zhang J. Role of the Cation-Chloride-Cotransporters in Cardiovascular Disease. Cells 2020; 9:2293. [PMID: 33066544 PMCID: PMC7602155 DOI: 10.3390/cells9102293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 02/05/2023] Open
Abstract
The SLC12 family of cation-chloride-cotransporters (CCCs) is comprised of potassium chloride cotransporters (KCCs), which mediate Cl- extrusion and sodium-potassium chloride cotransporters (N[K]CCs), which mediate Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. The functions of CCCs influence a variety of physiological processes, many of which overlap with the pathophysiology of cardiovascular disease. Although not all of the cotransporters have been linked to Mendelian genetic disorders, recent studies have provided new insights into their functional role in vascular and renal cells in addition to their contribution to cardiovascular diseases. Particularly, an imbalance in potassium levels promotes the pathogenesis of atherosclerosis and disturbances in sodium homeostasis are one of the causes of hypertension. Recent findings suggest hypothalamic signaling as a key signaling pathway in the pathophysiology of hypertension. In this review, we summarize and discuss the role of CCCs in cardiovascular disease with particular emphasis on knowledge gained in recent years on NKCCs and KCCs.
Collapse
Affiliation(s)
- Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, Hatherly Laboratories, University of Exeter, Exeter EX4 4PS, UK;
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, Hatherly Laboratories, University of Exeter, Exeter EX4 4PS, UK;
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
| |
Collapse
|
31
|
Mesocortical BDNF signaling mediates antidepressive-like effects of lithium. Neuropsychopharmacology 2020; 45:1557-1566. [PMID: 32428928 PMCID: PMC7360776 DOI: 10.1038/s41386-020-0713-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022]
Abstract
Lithium has been used to treat major depressive disorder, yet the neural circuit mechanisms underlying this therapeutic effect remain unknown. Here, we demonstrated that the ventral tegmental area (VTA) dopamine (DA) neurons that project to the medial prefrontal cortex (mPFC), but not to nucleus accumbens (NAc), contributed to the antidepressive-like effects of lithium. Projection-specific electrophysiological recordings revealed that high concentrations of lithium increased firing rates in mPFC-, but not NAc-, projecting VTA DA neurons in mice treated with chronic unpredictable mild stress (CMS). In parallel, chronic administration of high-dose lithium in CMS mice restored the firing properties of mPFC-projecting DA neurons, and also rescued CMS-induced depressive-like behaviors. Nevertheless, chronic lithium treatment was insufficient to change the basal firing rates in NAc-projecting VTA DA neurons. Furthermore, chemogenetic activation of mPFC-, but not NAc-, projecting VTA DA neurons mimicked the antidepressive-like effects of lithium in CMS mice. Chemogenetic downregulation of VTA-mPFC DA neurons' firing activity abolished the antidepressive-like effects of lithium in CMS mice. Finally, we found that the antidepressant-like effects induced by high-dose lithium were mediated by BNDF signaling in the mesocortical DA circuit. Together, these results demonstrated the role of mesocortical DA projection in antidepressive-like effects of lithium and established a circuit foundation for lithium-based antidepressive treatment.
Collapse
|
32
|
Wu HB, Shao K, Wang YC, Wang XC, Liu HL, Xie YT, Du RP. Research progress of CA125 and BDNF in serum of patients with acute myocardial infarction for predicting acute heart failure. Clin Hemorheol Microcirc 2020; 75:99-106. [PMID: 31868662 DOI: 10.3233/ch-190738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Hai-Bo Wu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Kai Shao
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yun-Can Wang
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xue-Chao Wang
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Hui-Liang Liu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yue-Tao Xie
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Rong-Pin Du
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
33
|
Hasegawa Y, Cheng C, Hayashi K, Takemoto Y, Kim-Mitsuyama S. Anti-apoptotic effects of BDNF-TrkB signaling in the treatment of hemorrhagic stroke. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2020.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
34
|
Japundžić-Žigon N, Lozić M, Šarenac O, Murphy D. Vasopressin & Oxytocin in Control of the Cardiovascular System: An Updated Review. Curr Neuropharmacol 2020; 18:14-33. [PMID: 31544693 PMCID: PMC7327933 DOI: 10.2174/1570159x17666190717150501] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/03/2019] [Accepted: 07/06/2019] [Indexed: 01/19/2023] Open
Abstract
Since the discovery of vasopressin (VP) and oxytocin (OT) in 1953, considerable knowledge has been gathered about their roles in cardiovascular homeostasis. Unraveling VP vasoconstrictor properties and V1a receptors in blood vessels generated powerful hemostatic drugs and drugs effective in the treatment of certain forms of circulatory collapse (shock). Recognition of the key role of VP in water balance via renal V2 receptors gave birth to aquaretic drugs found to be useful in advanced stages of congestive heart failure. There are still unexplored actions of VP and OT on the cardiovascular system, both at the periphery and in the brain that may open new venues in treatment of cardiovascular diseases. After a brief overview on VP, OT and their peripheral action on the cardiovascular system, this review focuses on newly discovered hypothalamic mechanisms involved in neurogenic control of the circulation in stress and disease.
Collapse
Affiliation(s)
| | - Maja Lozić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Šarenac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - David Murphy
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
35
|
Balapattabi K, Little JT, Bachelor ME, Cunningham RL, Cunningham JT. Sex Differences in the Regulation of Vasopressin and Oxytocin Secretion in Bile Duct-Ligated Rats. Neuroendocrinology 2020; 111:237-248. [PMID: 32335554 PMCID: PMC7584765 DOI: 10.1159/000508104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Hyponatremia due to elevated arginine vasopressin (AVP) secretion increases mortality in liver failure patients. No previous studies have addressed sex differences in hyponatremia in liver failure animal models. OBJECTIVE This study addressed this gap in our understanding of the potential sex differences in hyponatremia associated with increased AVP secretion. METHODS This study tested the role of sex in the development of hyponatremia using adult male, female, and ovariectomized (OVX) female bile duct-ligated (BDL) rats. RESULTS All BDL rats had significantly increased liver to body weight ratios compared to sham controls. Male BDL rats had hyponatremia with significant increases in plasma copeptin and FosB expression in supraoptic AVP neurons compared to male shams (all p < 0.05; 5-7). Female BDL rats did not become hyponatremic or demonstrate increased supraoptic AVP neuron activation and copeptin secretion compared to female shams. Plasma oxytocin was significantly higher in female BDL rats compared to female sham (p < 0.05; 6-10). This increase was not observed in male BDL rats. Ovariectomy significantly decreased plasma estradiol in sham rats compared to intact female sham (p < 0.05; 6-10). However, circulating estradiol was significantly elevated in OVX BDL rats compared to the OVX and female shams (p < 0.05; 6-10). Adrenal estradiol, testosterone, and dehydroepiandrosterone (DHEA) were measured to identify a possible source of circulating estradiol in OVX BDL rats. The OVX BDL rats had significantly increased adrenal estradiol along with significantly decreased adrenal testosterone and DHEA compared to OVX shams (all p < 0.05; 6-7). Plasma osmolality, hematocrit, copeptin, and AVP neuron activation were not significantly different between OVX BDL and OVX shams. Plasma oxytocin was significantly higher in OVX BDL rats compared to OVX sham. CONCLUSIONS Our results show that unlike male BDL rats, female and OVX BDL rats did not develop hyponatremia, supraoptic AVP neuron activation, or increased copeptin secretion compared to female shams. Adrenal estradiol might have compensated for the lack of ovarian estrogens in OVX BDL rats.
Collapse
Affiliation(s)
- Kirthikaa Balapattabi
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Joel T Little
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Martha E Bachelor
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA,
| |
Collapse
|
36
|
Unique Organization of Actin Cytoskeleton in Magnocellular Vasopressin Neurons in Normal Conditions and in Response to Salt-Loading. eNeuro 2020; 7:ENEURO.0351-19.2020. [PMID: 32209611 PMCID: PMC7189486 DOI: 10.1523/eneuro.0351-19.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/04/2022] Open
Abstract
Magnocellular neurosecretory cells (MNCs) are intrinsically osmosensitive and can be activated by increases in blood osmolality, triggering the release of antidiuretic hormone vasopressin (VP) to promote water retention. Hence, the activity of magnocellular VP neurons is one of the key elements contributing to the regulation of body fluid homeostasis in healthy organisms. Chronic exposure to high dietary salt leads to excessive activation of VP neurons, thereby elevating levels of circulating VP, which can cause increases in blood pressure contributing to salt-dependent hypertension. However, the molecular basis underlying high-salt diet-induced hyperactivation of magnocellular VP neurons remains not fully understood. Previous studies suggest that magnocellular neurosecretory neurons contain a subcortical layer of actin filaments and pharmacological stabilization of this actin network potentiates osmotically-induced activation of magnocellular neurons. Using super-resolution imaging in situ, we investigated the organization of the actin cytoskeleton in rat MNCs under normal physiological conditions and after a chronic increase in blood osmolality following 7 d of salt-loading (SL). We found that, in addition to the subcortical layer of actin filaments, magnocellular VP neurons are endowed with a unique network of cytoplasmic actin filaments throughout their somata. Moreover, we revealed that the density of both subcortical and cytoplasmic actin networks in magnocellular VP neurons is dramatically increased following SL. These results suggest that increased osmo-responsiveness of VP neurons following chronic exposure to high dietary salt may be mediated by the modulation of unique actin networks in magnocellular VP neurons, possibly contributing to elevated blood pressure in this condition.
Collapse
|
37
|
Tasker JG, Prager-Khoutorsky M, Teruyama R, Lemos JR, Amstrong WE. Advances in the neurophysiology of magnocellular neuroendocrine cells. J Neuroendocrinol 2020; 32:e12826. [PMID: 31917875 PMCID: PMC7192795 DOI: 10.1111/jne.12826] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Hypothalamic magnocellular neuroendocrine cells have unique electrical properties and a remarkable capacity for morphological and synaptic plasticity. Their large somatic size, their relatively uniform and dense clustering in the supraoptic and paraventricular nuclei, and their large axon terminals in the neurohypophysis make them an attractive target for direct electrophysiological interrogation. Here, we provide a brief review of significant recent findings in the neuroplasticity and neurophysiological properties of these neurones that were presented at the symposium "Electrophysiology of Magnocellular Neurons" during the 13th World Congress on Neurohypophysial Hormones in Ein Gedi, Israel in April 2019. Magnocellular vasopressin (VP) neurones respond directly to hypertonic stimulation with membrane depolarisation, which is triggered by cell shrinkage-induced opening of an N-terminal-truncated variant of transient receptor potential vanilloid type-1 (TRPV1) channels. New findings indicate that this mechanotransduction depends on actin and microtubule cytoskeletal networks, and that direct coupling of the TRPV1 channels to microtubules is responsible for mechanical gating of the channels. Vasopressin neurones also respond to osmostimulation by activation of epithelial Na+ channels (ENaC). It was shown recently that changes in ENaC activity modulate magnocellular neurone basal firing by generating tonic changes in membrane potential. Both oxytocin and VP neurones also undergo robust excitatory synapse plasticity during chronic osmotic stimulation. Recent findings indicate that new glutamate synapses induced during chronic salt loading express highly labile Ca2+ -permeable GluA1 receptors requiring continuous dendritic protein synthesis for synapse maintenance. Finally, recordings from the uniquely tractable neurohypophysial terminals recently revealed an unexpected property of activity-dependent neuropeptide release. A significant fraction of the voltage-dependent neurohypophysial neurosecretion was found to be independent of Ca2+ influx through voltage-gated Ca2+ channels. Together, these findings provide a snapshot of significant new advances in the electrophysiological signalling mechanisms and neuroplasticity of the hypothalamic-neurohypophysial system, a system that continues to make important contributions to the field of neurophysiology.
Collapse
Affiliation(s)
- Jeffrey G. Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Correspondence: Jeffrey Tasker, PhD, Tulane University, Cell and Molecular Biology Dept, 2000 Percival Stern Hall, New Orleans, LA 70118, USA; .; William Armstrong, PhD, University of Tennessee Health Science Center, Anatomy and Neurobiology Dept and Neuroscience Institute, 855 Monroe Ave, Memphis, TN 38163, USA;
| | | | - Ryoichi Teruyama
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - José R. Lemos
- Department of Microbiology and Physiological Systems & Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA, USA
| | - William E. Amstrong
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
- Correspondence: Jeffrey Tasker, PhD, Tulane University, Cell and Molecular Biology Dept, 2000 Percival Stern Hall, New Orleans, LA 70118, USA; .; William Armstrong, PhD, University of Tennessee Health Science Center, Anatomy and Neurobiology Dept and Neuroscience Institute, 855 Monroe Ave, Memphis, TN 38163, USA;
| |
Collapse
|
38
|
Mueller PJ, Fyk-Kolodziej BE, Azar TA, Llewellyn-Smith IJ. Subregional differences in GABA A receptor subunit expression in the rostral ventrolateral medulla of sedentary versus physically active rats. J Comp Neurol 2020; 528:1053-1075. [PMID: 31642070 PMCID: PMC7046220 DOI: 10.1002/cne.24798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022]
Abstract
Neurons in the rostral ventrolateral medulla (RVLM) regulate blood pressure through direct projections to spinal sympathetic preganglionic neurons. Only some RVLM neurons are active under resting conditions due to significant, tonic inhibition by gamma-aminobutyric acid (GABA). Withdrawal of GABAA receptor-mediated inhibition of the RVLM increases sympathetic outflow and blood pressure substantially, providing a mechanism by which the RVLM could contribute chronically to cardiovascular disease (CVD). Here, we tested the hypothesis that sedentary conditions, a major risk factor for CVD, increase GABAA receptors in RVLM, including its rostral extension (RVLMRE ), both of which contain bulbospinal catecholamine (C1) and non-C1 neurons. We examined GABAA receptor subunits GABAAα1 and GABAAα2 in the RVLM/RVLMRE of sedentary or physically active (10-12 weeks of wheel running) rats. Western blot analyses indicated that sedentary rats had lower expression of GABAAα1 and GABAAα2 subunits in RVLM but only GABAAα2 was lower in the RVLMRE of sedentary rats. Sedentary rats had significantly reduced expression of the chloride transporter, KCC2, suggesting less effective GABA-mediated inhibition compared to active rats. Retrograde tracing plus triple-label immunofluorescence identified fewer bulbospinal non-C1 neurons immunoreactive for GABAAα1 but a higher percentage of bulbospinal C1 neurons immunoreactive for GABAAα1 in sedentary animals. Sedentary conditions did not significantly affect the number of bulbospinal C1 or non-C1 neurons immunoreactive for GABAAα2 . These results suggest a complex interplay between GABAA receptor expression by spinally projecting C1 and non-C1 neurons and sedentary versus physically active conditions. They also provide plausible mechanisms for both enhanced sympathoexcitatory and sympathoinhibitory responses following sedentary conditions.
Collapse
Affiliation(s)
- Patrick J. Mueller
- Department of Physiology, Wayne State University School of
Medicine, Detroit MI
| | | | - Toni A. Azar
- Department of Physiology, Wayne State University School of
Medicine, Detroit MI
| | - Ida J. Llewellyn-Smith
- Department of Physiology, Wayne State University School of
Medicine, Detroit MI
- Cardiovascular Medicine, Human Physiology and Centre for
Neuroscience, College of Medicine and Public Health, Flinders University, Bedford
Park SA, AUSTRALIA
| |
Collapse
|
39
|
Matuska R, Zelena D, Könczöl K, Papp RS, Durst M, Guba D, Török B, Varnai P, Tóth ZE. Colocalized neurotransmitters in the hindbrain cooperate in adaptation to chronic hypernatremia. Brain Struct Funct 2020; 225:969-984. [PMID: 32200401 PMCID: PMC7166202 DOI: 10.1007/s00429-020-02049-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 02/13/2020] [Indexed: 11/29/2022]
Abstract
Chronic hypernatremia activates the central osmoregulatory mechanisms and inhibits the function of the hypothalamic-pituitary-adrenal (HPA) axis. Noradrenaline (NE) release into the periventricular anteroventral third ventricle region (AV3V), the supraoptic (SON) and hypothalamic paraventricular nuclei (PVN) from efferents of the caudal ventrolateral (cVLM) and dorsomedial (cDMM) medulla has been shown to be essential for the hypernatremia-evoked responses and for the HPA response to acute restraint. Notably, the medullary NE cell groups highly coexpress prolactin-releasing peptide (PrRP) and nesfatin-1/NUCB2 (nesfatin), therefore, we assumed they contributed to the reactions to chronic hypernatremia. To investigate this, we compared two models: homozygous Brattleboro rats with hereditary diabetes insipidus (DI) and Wistar rats subjected to chronic high salt solution (HS) intake. HS rats had higher plasma osmolality than DI rats. PrRP and nesfatin mRNA levels were higher in both models, in both medullary regions compared to controls. Elevated basal tyrosine hydroxylase (TH) expression and impaired restraint-induced TH, PrRP and nesfatin expression elevations in the cVLM were, however, detected only in HS, but not in DI rats. Simultaneously, only HS rats exhibited classical signs of chronic stress and severely blunted hormonal reactions to acute restraint. Data suggest that HPA axis responsiveness to restraint depends on the type of hypernatremia, and on NE capacity in the cVLM. Additionally, NE and PrRP signalization primarily of medullary origin is increased in the SON, PVN and AV3V in HS rats. This suggests a cooperative action in the adaptation responses and designates the AV3V as a new site for PrRP's action in hypernatremia.
Collapse
Affiliation(s)
- Rita Matuska
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Dóra Zelena
- Behavioral Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Katalin Könczöl
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Rege Sugárka Papp
- Human Brain Tissue Bank and Microdissection Laboratory, Semmelweis University, Budapest, Hungary
| | - Máté Durst
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Dorina Guba
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Bibiana Török
- Behavioral Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- Janos Szentagothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Peter Varnai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna E Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
40
|
Hicks AI, Barad Z, Sobrero A, Lean G, Jacob-Tomas S, Yang J, Choe KY, Prager-Khoutorsky M. Effects of salt loading on the organisation of microtubules in rat magnocellular vasopressin neurones. J Neuroendocrinol 2020; 32:e12817. [PMID: 31778225 DOI: 10.1111/jne.12817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/25/2022]
Abstract
Magnocellular vasopressin (VP) neurones are activated by increases in blood osmolality, leading to the secretion of VP into the circulation to promote water retention in the kidney, thus constituting a key mechanism for the regulation of body fluid homeostasis. However, chronic high salt intake can lead to excessive activation of VP neurones and increased circulating levels of VP, contributing to an elevation in blood pressure. Multiple extrinsic factors, such as synaptic inputs and glial cells, modulate the activity of VP neurones. Moreover, magnocellular neurones are intrinsically osmosensitive, and are activated by hypertonicity in the absence of neighbouring cells or synaptic contacts. Hypertonicity triggers cell shrinking, leading to the activation of VP neurones. This cell-autonomous activation is mediated by a scaffold of dense somatic microtubules, uniquely present in VP magnocellular neurones. Treating isolated magnocellular neurones with drugs modulating microtubule stability modifies the sensitivity of neuronal activation in response to acute hypertonic stimuli. However, whether the microtubule network is altered in conditions associated with enhanced neuronal activation and increased VP release, such as chronic high salt intake, remains unknown. We examined the organisation of microtubules in VP neurones of the supraoptic and paraventricular hypothalamic nuclei (SON and PVN, respectively) of rats subjected to salt-loading (drinking 2% NaCl for 7 days). Using super-resolution imaging, we found that the density of microtubules in magnocellular VP neurones from the SON and PVN was significantly increased, whereas the density and organisation of microtubules remain unchanged in other hypothalamic neurones, as well as in neurones from other brain areas (e.g., hippocampus, cortex). We propose that the increase in microtubule density in magnocellular VP neurones in salt-loading promotes their enhanced activation, possibly contributing to elevated blood pressure in this condition.
Collapse
Affiliation(s)
| | - Zsuzsanna Barad
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Alberto Sobrero
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Graham Lean
- Department of Physiology, McGill University, Montreal, QC, Canada
| | | | - Jieyi Yang
- Department of Physiology, McGill University, Montreal, QC, Canada
| | | | | |
Collapse
|
41
|
He D, Chen H, Zeng M, Xia C, Wang J, Shen L, Zhu D, Chen Y, Wang J. Asthmatic Airway Vagal Hypertonia Involves Chloride Dyshomeostasis of Preganglionic Neurons in Rats. Front Neurosci 2020; 14:31. [PMID: 32082109 PMCID: PMC7005078 DOI: 10.3389/fnins.2020.00031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 01/10/2020] [Indexed: 11/17/2022] Open
Abstract
Airway vagal hypertonia is closely related to the severity of asthma; however, the mechanisms of its genesis are unclear. This study aims to prove that asthmatic airway vagal hypertonia involves neuronal Cl– dyshomeostasis. The experimental airway allergy model was prepared with ovalbumin in male adult Sprague-Dawley rats. Plethysmography was used to evaluate airway vagal response to intracisternally injected γ-aminobutyric acid (GABA). Immunofluorescent staining and Western-blot assay were used to examine the expression of microglia-specific proteins, Na+-K+-2Cl– co-transporter 1 (NKCC1), K+-Cl– co-transporter 2 (KCC2) and brain-derived nerve growth factor (BDNF) in airway vagal centers. Pulmonary inflammatory changes were examined with hematoxylin and eosin staining of lung sections and ELISA assay of ovalbumin-specific IgE in bronchoalveolar lavage fluid (BALF). The results showed that histochemically, experimental airway allergy activated microglia, upregulated NKCC1, downregulated KCC2, and increased the content of BDNF in airway vagal centers. Functionally, experimental airway allergy augmented the excitatory airway vagal response to intracisternally injected GABA, which was attenuated by intracisternally pre-injected NKCC1 inhibitor bumetanide. All of the changes induced by experimental airway allergy were prevented or mitigated by chronic intracerebroventricular or intraperitoneal injection of minocycline, an inhibitor of microglia activation. These results demonstrate that experimental airway allergy augments the excitatory response of airway vagal centers to GABA, which might be the result of neuronal Cl– dyshomeostasis subsequent to microglia activation, increased BDNF release and altered expression of Cl– transporters. Cl– dyshomeostasis in airway vagal centers might contribute to the genesis of airway vagal hypertonia in asthma.
Collapse
Affiliation(s)
- Ding He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ming Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Danian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yonghua Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jijiang Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Vasopressin inactivation: Role of insulin-regulated aminopeptidase. VITAMINS AND HORMONES 2019; 113:101-128. [PMID: 32138946 DOI: 10.1016/bs.vh.2019.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The physiological importance of vasopressin inactivation has long been appreciated, but the mechanisms and potential pathophysiologic roles of this process remain active subjects of research. Human Placental Leucine Aminopeptidase (P-LAP, encoded by the LNPEP gene) is an important determinant of vasopressinase activity during pregnancy and is associated with gestational diabetes insipidus and preeclampsia. Insulin-Regulated Aminopeptidase (IRAP), the rodent homologue of P-LAP, is coregulated with the insulin-responsive glucose transporter, GLUT4, in adipose and muscle cells. Recently, the Tether containing a UBX domain for GLUT4 (TUG) protein was shown to mediate the coordinated regulation of water and glucose homeostasis. TUG sequesters IRAP and GLUT4 intracellularly in the absence of insulin. Insulin and other stimuli cause the proteolytic cleavage of TUG to mobilize these proteins to the cell surface, where IRAP acts to terminate the activity of circulating vasopressin. Intriguingly, genetic variation in LNPEP is associated with the vasopressin response and mortality during sepsis, and increased copeptin, a marker of vasopressin secretion, is associated with cardiovascular and metabolic disease. We propose that in the setting of insulin resistance in muscle, increased cell-surface IRAP and accelerated vasopressin degradation cause a compensatory increase in vasopressin secretion. The increased vasopressin concentrations present at the kidneys then contribute to hypertension in the metabolic syndrome. Further analyses of metabolism and of vasopressin and copeptin may yield novel insights into a unified pathophysiologic mechanism linking insulin resistance and hypertension, and potentially other components of the metabolic syndrome, in humans.
Collapse
|
43
|
Thorsdottir D, Cruickshank NC, Einwag Z, Hennig GW, Erdos B. BDNF downregulates β-adrenergic receptor-mediated hypotensive mechanisms in the paraventricular nucleus of the hypothalamus. Am J Physiol Heart Circ Physiol 2019; 317:H1258-H1271. [PMID: 31603352 DOI: 10.1152/ajpheart.00478.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is upregulated in the paraventricular nucleus of the hypothalamus (PVN) in response to hypertensive stimuli such as stress and hyperosmolality, and BDNF acting in the PVN plays a key role in elevating sympathetic activity and blood pressure. However, downstream mechanisms mediating these effects remain unclear. We tested the hypothesis that BDNF increases blood pressure, in part by diminishing inhibitory hypotensive input from nucleus of the solitary tract (NTS) catecholaminergic neurons projecting to the PVN. Male Sprague-Dawley rats received bilateral PVN injections of viral vectors expressing either green fluorescent protein (GFP) or BDNF and bilateral NTS injections of vehicle or anti-dopamine-β-hydroxylase-conjugated saporin (DSAP), a neurotoxin that selectively lesions noradrenergic and adrenergic neurons. BDNF overexpression in the PVN without NTS lesioning significantly increased mean arterial pressure (MAP) in awake animals by 18.7 ± 1.8 mmHg. DSAP treatment also increased MAP in the GFP group, by 9.8 ± 3.2 mmHg, but failed to affect MAP in the BDNF group, indicating a BDNF-induced loss of NTS catecholaminergic hypotensive effects. In addition, in α-chloralose-urethane-anesthetized rats, hypotensive responses to PVN injections of the β-adrenergic agonist isoprenaline were significantly attenuated by BDNF overexpression, whereas PVN injections of phenylephrine had no effect on blood pressure. BDNF treatment was also found to significantly reduce β1-adrenergic receptor mRNA expression in the PVN, whereas expression of other adrenergic receptors was unaffected. In summary, increased BDNF expression in the PVN elevates blood pressure, in part by downregulating β-receptor signaling and diminishing hypotensive catecholaminergic input from the NTS to the PVN.NEW & NOTEWORTHY We have shown that BDNF, a key hypothalamic regulator of blood pressure, disrupts catecholaminergic signaling between the NTS and the PVN by reducing the responsiveness of PVN neurons to inhibitory hypotensive β-adrenergic input from the NTS. This may be occurring partly via BDNF-mediated downregulation of β1-adrenergic receptor expression in the PVN and results in an increase in blood pressure.
Collapse
Affiliation(s)
| | | | - Zachary Einwag
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Grant W Hennig
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| |
Collapse
|
44
|
Balapattabi K, Little JT, Bachelor M, Cunningham JT. Brain-Derived Neurotrophic Factor and Supraoptic Vasopressin Neurons in Hyponatremia. Neuroendocrinology 2019; 110:630-641. [PMID: 31557760 PMCID: PMC7385921 DOI: 10.1159/000503723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022]
Abstract
Hyponatremia due to elevated arginine vasopressin (AVP) secretion increases mortality in liver failure patients. The mechanisms causing dysregulation of AVP secretion are unknown. Our hypothesis is that inappropriate AVP release associated with liver failure is due to increased brain-derived neurotrophic factor (BDNF) in the supraoptic nucleus (SON). BDNF diminishes GABAA inhibition in SON AVP neurons by increasing intracellular chloride through tyrosine receptor kinase B (TrkB) activation and downregulation of K+/Cl- cotransporter 2 (KCC2). This loss of inhibition could increase AVP secretion. This hypothesis was tested using shRNA against BDNF (shBDNF) in the SON in bile duct ligated (BDL) male rats. All BDL rats had significantly increased liver weight (p < 0.05; 6-9) compared to shams. BDL rats with control -shRNA injections (BDL scrambled [SCR]) developed hyponatremia with increased plasma AVP and copeptin (CPP; all p < 0.05; 6-9) compared to sham groups. This is the first study to show that phosphorylation of TrkB is significantly increased along with significant decrease in phosphorylation of KCC2 in BDL SCR rats compared to the sham rats (p < 0.05;6-8). Knockdown of BDNF in the SON of BDL rats (BDL shBDNF) significantly increased plasma osmolality and hematocrit compared to BDL SCR rats (p < 0.05; 6-9). The BDL shBDNF rats had significant (p < 0.05; 6-9) decreases in plasma AVP and CPP concentration compared to BDL SCR rats. The BDNF knockdown also significantly blocked the increase in TrkB phosphorylation and decrease in KCC2 phosphorylation (p < 0.05; 6-8). The results indicate that BDNF produced in the SON contributes to increased AVP secretion and hyponatremia during liver failure.
Collapse
Affiliation(s)
- Kirthikaa Balapattabi
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Joel T Little
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Martha Bachelor
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA,
| |
Collapse
|
45
|
Dong Y, Jan C, Zou Z, Dong B, Wang Z, Yang Z, Li Y, Wen B, Ma Y, Song Y, Ma J, Sawyer SM, Patton GC. Effect of Overweight and Obesity on High Blood Pressure in Chinese Children and Adolescents. Obesity (Silver Spring) 2019; 27:1503-1512. [PMID: 31328902 DOI: 10.1002/oby.22562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/20/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE This study aimed to compare the secular trends of high blood pressure (HBP) and the effects of overweight and obesity on HBP between Chinese ethnic minority and Han children and adolescents . METHODS Data were collected from 224,151 Chinese ethnic minority and 664,094 Han children and adolescents aged 7 to 18 years during three successive, national cross-sectional surveys (2005, 2010, and 2014). Logistic regression and population-attributable risk analyses were used to evaluate the association between HBP and overweight and obesity. RESULTS HBP prevalence in ethnic minorities increased from 4.8% in 2005 to 6.3% in 2014, which was significantly higher than the variable HBP trends (4.1% to 5.5%) in Han children and adolescents. Both ethnic minority and Han children and adolescents experienced a rapid increase in overweight and obesity, but the pace of growth for HBP, overweight, and obesity was faster in ethnic minorities than in their Han peers. Moreover, the effects of obesity on HBP in ethnic minorities showed a sustained increase over time but were stable for the Han. CONCLUSIONS Higher HBP prevalence, faster obesity increases, and a stronger impact of obesity on HBP in children and adolescents of Chinese ethnic minorities predict their looming burden of HBP, which suggests that attention to the cardiovascular disease risks in children and adolescents from ethnic minorities is indicated to reduce their future adult risk.
Collapse
Affiliation(s)
- Yanhui Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Catherine Jan
- George Institute for Global Health, School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Zhiyong Zou
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Bin Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Zhenghe Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Zhaogeng Yang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Yanhui Li
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Bo Wen
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Yinghua Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Yi Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Susan M Sawyer
- Faculty of Medicine, Department of Paediatrics, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria, Australia
| | - George C Patton
- Faculty of Medicine, Department of Paediatrics, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
46
|
Kim YB, Colwell CS, Kim YI. Long-term ionic plasticity of GABAergic signalling in the hypothalamus. J Neuroendocrinol 2019; 31:e12753. [PMID: 31166034 DOI: 10.1111/jne.12753] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 12/23/2022]
Abstract
The hypothalamus contains a number of nuclei that subserve a variety of functions, including generation of circadian rhythms, regulation of hormone secretion and maintenance of homeostatic levels for a variety of physiological parameters. Within the hypothalamus, γ-amino-butyric acid (GABA) is one of the major neurotransmitters responsible for cellular communication. Although GABA most commonly serves as an inhibitory neurotransmitter, a growing body of evidence indicates that it can evoke post-synaptic excitation as a result of the active regulation of intracellular chloride concentration. In this review, we consider the evidence for this ionic plasticity of GABAergic synaptic transmission in five distinct cases in hypothalamic cell populations. We argue that this plasticity serves as part of the functional response to or is at least associated with dehydration, lactation, hypertension and stress. As such, GABA excitation should be considered as part of the core homeostatic mechanisms of the hypothalamus.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University, Seoul, Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, UCLA School of Medicine, Los Angeles, CA, USA
| | - Yang In Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University, Seoul, Korea
| |
Collapse
|
47
|
Balapattabi K, Farmer GE, Knapp BA, Little JT, Bachelor M, Yuan JP, Cunningham JT. Effects of salt-loading on supraoptic vasopressin neurones assessed by ClopHensorN chloride imaging. J Neuroendocrinol 2019; 31:e12752. [PMID: 31136029 PMCID: PMC7041405 DOI: 10.1111/jne.12752] [Citation(s) in RCA: 9] [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: 03/07/2019] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022]
Abstract
Salt-loading (SL) impairs GABAA inhibition of arginine vasopressin (AVP) neurones in the supraoptic nucleus (SON) of the hypothalamus. Based on previous studies, we hypothesised that SL activates tyrosine receptor kinase B (TrkB), down-regulating the activity of K+ /Cl- co-transporter2 (KCC2) and up-regulating Na+ /K+ /Cl- co-transporter1 (NKCC1). These changes in chloride transport would result in increased [Cl- ]i in SON AVP neurones. The study combined virally-mediated chloride imaging with ClopHensorN with a single-cell western blot analysis. An adeno-associated virus with ClopHensorN and a vasopressin promoter (AAV2-0VP1-ClopHensorN) was bilaterally injected in the SON of adult male Sprague-Dawley rats that were either euhydrated (Eu) or salt-loaded (SL) for 7 days. Acutely dissociated SON neurones expressing ClopHensorN were tested for decreases or increases in [Cl- ]i in response to focal application of the GABAA agonist muscimol (100 μmol L-1 ). SON AVP neurones from Eu rats showed muscimol-induced chloride influx (P < 0.05;23/35). SON AVP neurones from SL rats either significantly increased chloride efflux (P < 0.05;27/39) or did not change chloride flux (12/39). The SON AVP neurones that responded to muscimol appeared to be viable and expressed KCC2 and β-actin. Neurones that did not respond during chloride imaging did not show KCC2 and β-actin protein expression. The KCC2 antagonist (VU0240551,10 μmol L-1 ) significantly blocked the chloride influx in cells from Eu rats but did not affect cells from SL rats. A NKCC1 antagonist (bumetanide,10 μmol L-1 ) significantly blocked the chloride efflux in cells from SL rats but had no effect on cells from Eu rats. Blocking NKCC1 using bumetanide had less of an effect on the muscimol-induced Cl- influx in Eu rat neurones compared to the KCC2 antagonist. The TrkB antagonist (AnA-12) (50 μmol L-1 ) and protein kinase inhibitor (K252a) (100 nmol L-1 ) each significantly blocked chloride efflux in SON AVP neurones from SL rats. Salt-loading increases [Cl- ]i in SON AVP neurones via a TrKB-KCC2-NKCC1-dependent mechanism in rats.
Collapse
Affiliation(s)
- Kirthikaa Balapattabi
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - Blayne A Knapp
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - Joel T Little
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - Martha Bachelor
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - Joseph P Yuan
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas
| |
Collapse
|
48
|
Di S, Jiang Z, Wang S, Harrison LM, Castro-Echeverry E, Stuart TC, Wolf ME, Tasker JG. Labile Calcium-Permeable AMPA Receptors Constitute New Glutamate Synapses Formed in Hypothalamic Neuroendocrine Cells during Salt Loading. eNeuro 2019; 6:ENEURO.0112-19.2019. [PMID: 31300543 PMCID: PMC6675872 DOI: 10.1523/eneuro.0112-19.2019] [Citation(s) in RCA: 6] [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: 03/22/2019] [Revised: 06/13/2019] [Accepted: 06/21/2019] [Indexed: 11/21/2022] Open
Abstract
Magnocellular neuroendocrine cells (MNCs) of the hypothalamus play a critical role in the regulation of fluid and electrolyte homeostasis. They undergo a dramatic structural and functional plasticity under sustained hyperosmotic conditions, including an increase in afferent glutamatergic synaptic innervation. We tested for a postulated increase in glutamate AMPA receptor expression and signaling in magnocellular neurons of the male rat hypothalamic supraoptic nucleus (SON) induced by chronic salt loading. While without effect on GluA1-4 subunit mRNA, salt loading with 2% saline for 5-7 d resulted in a selective increase in AMPA receptor GluA1 protein expression in the SON, with no change in GluA2-4 protein expression, suggesting an increase in the ratio of GluA1 to GluA2 subunits. Salt loading induced a corresponding increase in EPSCs in both oxytocin (OT) and vasopressin (VP) neurons, with properties characteristic of calcium-permeable AMPA receptor-mediated currents. Unexpectedly, the emergent AMPA synaptic currents were silenced by blocking protein synthesis and mammalian target of rapamycin (mTOR) activity in the slices, suggesting that the new glutamate synapses induced by salt loading require continuous dendritic protein synthesis for maintenance. These findings indicate that chronic salt loading leads to the induction of highly labile glutamate synapses in OT and VP neurons that are comprised of calcium-permeable homomeric GluA1 AMPA receptors. The glutamate-induced calcium influx via calcium-permeable AMPA receptors would be expected to play a key role in the induction and/or maintenance of activity-dependent synaptic plasticity that occurs in the magnocellular neurons during chronic osmotic stimulation.
Collapse
Affiliation(s)
- Shi Di
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - ZhiYing Jiang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Sen Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Laura M Harrison
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | | | - Thomas C Stuart
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Marina E Wolf
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
- Tulane Brain Institute, Tulane University, New Orleans, LA 70118
| |
Collapse
|
49
|
Ivetic M, Bhattacharyya A, Zemkova H. P2X2 Receptor Expression and Function Is Upregulated in the Rat Supraoptic Nucleus Stimulated Through Refeeding After Fasting. Front Cell Neurosci 2019; 13:284. [PMID: 31297050 PMCID: PMC6607214 DOI: 10.3389/fncel.2019.00284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/12/2019] [Indexed: 01/12/2023] Open
Abstract
Magnocellular neurons in the supraoptic nucleus (SON), which synthesize and release arginine vasopressin (AVP) and oxytocin (OT), express several subtypes of ATP-stimulated purinergic P2X receptors (P2XR) that modulate neuronal activity as well as neurotransmitter and hormone release. However, the physiological impact of this modulation is not well understood. Here, we tested a hypothesis that P2XRs play a role in the sustained release of hormones from SON neurons stimulated through fasting/refeeding. We studied the effect of 2 h of refeeding after 48 h of fasting on P2XR and P2YR mRNA expression and ATP-induced presynaptic and postsynaptic responses in the SON of 30-day-old rats. Quantitative real-time PCR revealed that the expression of P2X2R and AVP mRNA was upregulated, whereas P2X4R, P2X7R, P2Y2R, and OT mRNA levels were not significantly changed and P2Y1R mRNA expression was decreased. Whole-cell patch clamp recordings performed on isolated rat brain slices showed that the amplitude of the ATP-stimulated somatic current and the ATP-induced increases in the frequency of spontaneous GABAergic inhibitory postsynaptic currents were significantly higher in SON neurons from fasted/refed rats than in SON neurons from normally fed rats. No evidence was found for changes in the presynaptic effect of ATP in SON neurons not expressing somatic P2XRs. These results suggest that the increased activity of SON neurons synthesizing AVP is associated with enhanced expression of P2X2Rs on neuronal cell bodies and their GABAergic presynaptic nerve terminals.
Collapse
Affiliation(s)
- Milorad Ivetic
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Anirban Bhattacharyya
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Hana Zemkova
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| |
Collapse
|
50
|
Dos-Santos RC, Reis LC, Perello M, Ferguson AV, Mecawi AS. The actions of ghrelin in the paraventricular nucleus: energy balance and neuroendocrine implications. Ann N Y Acad Sci 2019; 1455:81-97. [PMID: 31008525 DOI: 10.1111/nyas.14087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/28/2019] [Accepted: 03/10/2019] [Indexed: 12/15/2022]
Abstract
Ghrelin is a peptide mainly produced and secreted by the stomach. Since its discovery, the impact of ghrelin on the regulation of food intake has been the most studied function of this hormone; however, ghrelin affects a wide range of physiological systems, many of which are controlled by the hypothalamic paraventricular nucleus (PVN). Several pathways may mediate the effects of ghrelin on PVN neurons, such as direct or indirect effects mediated by circumventricular organs and/or the arcuate nucleus. The ghrelin receptor is expressed in PVN neurons, and the peripheral or intracerebroventricular administration of ghrelin affects PVN neuronal activity. Intra-PVN application of ghrelin increases food intake and decreases fat oxidation, which chronically contribute to the increased adiposity. Additionally, ghrelin modulates the neuroendocrine axes controlled by the PVN, increasing the release of vasopressin and oxytocin by magnocellular neurons and corticotropin-releasing hormone by neuroendocrine parvocellular neurons, while possibly inhibiting the release of thyrotropin-releasing hormone. Thus, the PVN is an important target for the actions of ghrelin. Our review discusses the mechanisms of ghrelin actions in the PVN, and its potential implications for energy balance, neuroendocrine, and integrative physiological control.
Collapse
Affiliation(s)
- Raoni C Dos-Santos
- Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Luís C Reis
- Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology, La Plata, Argentina
| | - Alastair V Ferguson
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - André S Mecawi
- Laboratory of Neuroendocrinology, Department of Biophysics, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|