1
|
Gao HL, Yang Y, Tian H, Fu LY, Liu KL, Jia XY, Shi XL, Kang YM, Yu XJ. Inhibition of CB1R in the Hypothalamic Paraventricular Nucleus Ameliorates Hypertension Through Wnt/β-Catenin/RAS Pathway. Cardiovasc Toxicol 2025; 25:9-23. [PMID: 39467886 DOI: 10.1007/s12012-024-09938-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024]
Abstract
The hypothalamic paraventricular nucleus (PVN), as an important integrating center, plays a prominent role in the pathogenesis of hypertension, in maintaining the stability of cardiovascular activity through peripheral sympathetic nervous activity and secretion of various humoral factors. Acknowledging that the mechanistic targets of the endocannabinoid type 1 receptor (CB1R) are the key signaling systems involved in the regulation of hypertension, we sought to clarify whether inhibition of CB1R within the PVN ameliorates hypertension through Wnt/β-catenin/RAS pathway. Spontaneously hypertensive rats (SHRs) and Wistar Kyoto rats were randomly assigned to different groups and treated with bilateral PVN injections of AM251 (CB1R antagonist, 10 µg/h) or vehicle (artificial cerebrospinal fluid, aCSF) for four weeks. Bilateral PVN injections of AM251 significantly decreased the heart rate, the body weight and the mean arterial pressure in SHRs. AM251 lowered the expression of CB1R, Wnt3, active-β-catenin, p-IKKβ, RAS components, pro-inflammatory cytokines and elevated the expression level of Glycogen synthase kinase3β and Superoxide Dismutase in the PVN of hypertensive rats. Our findings suggest that inhibition of CB1R in the PVN ameliorates hypertension through Wnt/β-catenin/RAS pathway and broaden our current understanding of the pathological mechanism and clinical treatment of hypertension.
Collapse
Affiliation(s)
- Hong-Li Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
| | - Yu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Hua Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
- Department of Diagnosis, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Li-Yan Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
| | - Kai-Li Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
| | - Xiu-Yue Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Xiao-Lian Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China.
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University, Ministry of Education, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
2
|
Nist KM, Bard H, McBride B, Capriglione AL, Moreira JD, Farb DH, Wainford RD. Losartan attenuates sex-dependent hypertension, neuroinflammation, and cognitive impairment in the aging male sprague-dawley rat. GeroScience 2024:10.1007/s11357-024-01409-4. [PMID: 39627570 DOI: 10.1007/s11357-024-01409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/22/2024] [Indexed: 01/18/2025] Open
Abstract
The prevalence of hypertension increases with age and is the leading modifiable risk factor for cognitive impairment and dementia. At present, the neural mechanisms promoting hypertension across the lifespan are incompletely understood. Using the Sprague-Dawley (SD) rat as a model of normal aging, we hypothesized (1) blood brain barrier (BBB) disruption and neuroinflammation in the paraventricular nucleus (PVN) of the hypothalamus enhances sympathetic tone and contributes to age-dependent hypertension, (2) age-dependent hypertension is associated with cognitive impairment, and (3) lowering blood pressure in aged rats with established hypertension improves cognitive function. We found male, but not female, rats develop age-dependent hypertension with enhanced sympathetic tone, BBB disruption, and neuroinflammation in the PVN. Aged hypertensive male rats also showed impairments in recognition and spatial memory. Utilizing pharmacological interventions, blood pressure was lowered in male rats with established hypertension using either losartan (LOS) or hydrochlorothiazide. However, only losartan improved recognition memory. Further, LOS reduced BBB disruption, microglial activation, astrocyte reactivity, and proinflammatory cytokine expression in the PVN which we speculate contributes to a decrease in blood pressure. These data show SD rats develop age-dependent hypertension and cognitive impairment in a sex-dependent manner. However, not all antihypertensive agents improve cognitive function equally as only losartan, an angiotensin II type 1 receptor antagonist (AT1R) improved recognition memory. Thus, AT1R antagonists represent a potential therapeutic approach for treating cognitive decline in the aging population.
Collapse
Affiliation(s)
- Kayla M Nist
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Whitaker Cardiovascular Institute, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Hannah Bard
- Whitaker Cardiovascular Institute, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Brannon McBride
- Whitaker Cardiovascular Institute, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Angela L Capriglione
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Whitaker Cardiovascular Institute, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jesse D Moreira
- Whitaker Cardiovascular Institute, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Health Sciences, Sargent College, Boston University, Boston, MA, USA
| | - David H Farb
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Richard D Wainford
- Whitaker Cardiovascular Institute, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N220, USA.
| |
Collapse
|
3
|
Frame AA, Nist KM, Kim K, Puleo F, Moreira JD, Swaldi H, McKenna J, Wainford RD. Integrated renal and sympathetic mechanisms underlying the development of sex- and age-dependent hypertension and the salt sensitivity of blood pressure. GeroScience 2024; 46:6435-6458. [PMID: 38976131 PMCID: PMC11494650 DOI: 10.1007/s11357-024-01266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Aging is a non-modifiable understudied risk factor for hypertension. We hypothesized that sympathetically mediated activation of renal sodium reabsorption drives age-dependent hypertension and the salt sensitivity of blood pressure (BP). Using 3-, 8-, and 16-month-old male and female Sprague-Dawley rats as a model of normal aging, we assessed BP, indices of sympathetic tone, and the physiological responses to acute and chronic sodium challenge including sodium chloride cotransporter (NCC) regulation. The effects of renal nerve ablation and NCC antagonism were assessed in hypertensive male rats. We observed sex-dependent impaired renal sodium handling (24 h sodium balance (meq), male 3-month 0.36 ± 0.1 vs. 16-month 0.84 ± 0.2; sodium load excreted during 5% bodyweight isotonic saline volume expansion (%) male 3-month 77 ± 5 vs. 16-month 22 ± 8), hypertension (MAP (mmHg) male 3-month 123 ± 4 vs. 16-month 148 ± 6), and the salt sensitivity of BP in aged male, but not female, rats. Attenuated sympathoinhibitory afferent renal nerve (ARN) responses contributed to increased sympathetic tone and hypertension in male rats. Increased sympathetic tone contributes to renal sodium retention, in part through increased NCC activity via a dysfunctional with-no-lysine kinase-(WNK) STE20/SPS1-related proline/alanine-rich kinase signaling pathway, to drive hypertension and the salt sensitivity of BP in aged male rats. NCC antagonism and renal nerve ablation, which reduced WNK dysfunction and decreased NCC activity, attenuated age-dependent hypertension in male Sprague-Dawley rats. The contribution of an impaired sympathoinhibitory ARN reflex to sex- and age-dependent hypertension in an NCC-dependent manner, via an impaired WNK1/WNK4 dynamic, suggests this pathway as a mechanism-based target for the treatment of age-dependent hypertension.
Collapse
Affiliation(s)
- Alissa A Frame
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kayla M Nist
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kiyoung Kim
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Franco Puleo
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse D Moreira
- Department of Health Sciences, Sargent College, Boston University, Boston, MA, USA
| | - Hailey Swaldi
- Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N22030322, USA
| | - James McKenna
- Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N22030322, USA
| | - Richard D Wainford
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N22030322, USA.
| |
Collapse
|
4
|
Althammer F, Roy RK, Kirchner MK, Podpecan Y, Helen J, McGrath S, Lira EC, Stern JE. Angiotensin-II drives changes in microglia-vascular interactions in rats with heart failure. Commun Biol 2024; 7:1537. [PMID: 39562706 PMCID: PMC11577102 DOI: 10.1038/s42003-024-07229-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Activation of microglia, the resident immune cells of the central nervous system, leading to the subsequent release of pro-inflammatory cytokines, has been linked to cardiac remodeling, autonomic disbalance, and cognitive deficits in heart failure (HF). While previous studies emphasized the role of hippocampal Angiotensin II (AngII) signaling in HF-induced microglial activation, unanswered mechanistic questions persist. Evidence suggests significant interactions between microglia and local microvasculature, potentially affecting blood-brain barrier integrity and cerebral blood flow regulation. Still, whether the microglial-vascular interface is affected in the brain during HF remains unknown. Using a well-established ischemic HF rat model, we demonstrate the increased abundance of vessel-associated microglia (VAM) in HF rat hippocampi, along with an increased expression of AngII AT1a receptors. Acute AngII administration to sham rats induced microglia recruitment to brain capillaries, along with increased expression of TNFα. Conversely, administering an AT1aR blocker to HF rats prevented the recruitment of microglia to blood vessels, normalizing their levels to those in healthy rats. These results highlight the critical importance of a rather understudied phenomenon (i.e., microglia-vascular interactions in the brain) in the context of the pathophysiology of a highly prevalent cardiovascular disease, and unveil novel potential therapeutic avenues aimed at mitigating neuroinflammation in cardiovascular diseases.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Matthew K Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Yuval Podpecan
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jemima Helen
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Shaina McGrath
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Elba Campos Lira
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA.
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
5
|
Liang YF, You QX, Chen SY, Ni L, Meng XL, Gao JX, Ren YB, Song HJ, Su JL, Teng Y, Gu QY, Lv C, Yuan BY, Wang X, Zheng YT, Zhang DD. The Impact of Hydrogen Sulfide in the Paraventricular Nucleus on the MAPK Pathway in High Salt-Induced Hypertension. J Cardiovasc Pharmacol 2024; 84:468-478. [PMID: 39115898 PMCID: PMC11446517 DOI: 10.1097/fjc.0000000000001622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 08/01/2024] [Indexed: 10/04/2024]
Abstract
ABSTRACT The hypothalamic paraventricular nucleus (PVN) plays a central role in regulating cardiovascular activity and blood pressure. We administered hydroxylamine hydrochloride (HA), a cystathionine-β-synthase inhibitor, into the PVN to suppress endogenous hydrogen sulfide and investigate its effects on the mitogen-activated protein kinase (MAPK) pathway in high salt (HS)-induced hypertension. We randomly divided 40 male Dahl salt-sensitive rats into 4 groups: the normal salt (NS) + PVN vehicle group, the NS + PVN HA group, the HS + PVN vehicle group, and the HS + PVN HA group, with 10 rats in each group. The rats in the NS groups were fed a NS diet containing 0.3% NaCl, while the HS groups were fed a HS diet containing 8% NaCl. The mean arterial pressure was calculated after noninvasive measurement using an automatic sphygmomanometer to occlude the tail cuff once a week. HA or vehicle was infused into the bilateral PVN using Alzet osmotic mini pumps for 6 weeks after the hypertension model was successfully established. We measured the levels of H 2 S in the PVN and plasma norepinephrine using enzyme linked immunosorbent assay. In addition, we assessed the parameters of the MAPK pathway, inflammation, and oxidative stress through western blotting, immunohistochemical analysis, or real-time polymerase chain reaction. In this study, we discovered that decreased levels of endogenous hydrogen sulfide in the PVN contributed to the onset of HS-induced hypertension. This was linked to the activation of the MAPK signaling pathway, proinflammatory cytokines, and oxidative stress in the PVN, as well as the activation of the sympathetic nervous system.
Collapse
Affiliation(s)
- Yan-Feng Liang
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Qing-Xin You
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Shu-Yue Chen
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Lei Ni
- First Affiliated Hospital, Jiamusi University, Jiamusi, China
| | - Xiang-Lian Meng
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, China
| | - Jian-Xiang Gao
- Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi, China; and
| | - Yong-Bo Ren
- First Affiliated Hospital, Jiamusi University, Jiamusi, China
| | - Han-Jun Song
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Jia-Lu Su
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Yang Teng
- School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Qing-Yun Gu
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Chao Lv
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Bo-Yang Yuan
- First Affiliated Hospital, Jiamusi University, Jiamusi, China
| | - Xuan Wang
- First Affiliated Hospital, Jiamusi University, Jiamusi, China
| | - Yong-Tai Zheng
- First Affiliated Hospital, Jiamusi University, Jiamusi, China
| | - Dong-Dong Zhang
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
6
|
Kim K, Nist KM, Puleo F, McKenna J, Wainford RD. Sex differences in dietary sodium evoked NCC regulation and blood pressure in male and female Sprague-Dawley, Dahl salt-resistant, and Dahl salt-sensitive rats. Am J Physiol Renal Physiol 2024; 327:F277-F289. [PMID: 38813592 PMCID: PMC11460337 DOI: 10.1152/ajprenal.00150.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
Hypertension affects approximately one in two United States adults and sex plays an important role in the pathogenesis of hypertension. The Na+-Cl- cotransporter (NCC), regulated by a kinase network including with-no-lysine kinase (WNK)1 and WNK4, STE20/SPS1-related proline alanine-rich kinase (SPAK), and oxidative stress response 1 (OxSR1), is critical to Na+ reabsorption and blood pressure regulation. Dietary salt differentially modulates NCC in salt-sensitive and salt-resistant rats, in part by modulation of WNK/SPAK/OxSR1 signaling. In this study, we tested the hypothesis that sex-dependent differences in NCC regulation contribute to the development of the salt sensitivity of blood pressure using male and female Sprague-Dawley (SD), Dahl salt-resistant (DSR), and Dahl salt-sensitive (DSS) rats. In normotensive salt-resistant SD and DSR rats, a high-salt diet evoked significant decreases in NCC activity, expression, and phosphorylation. In males, these changes were associated with no change in WNK1 expression, a decrease in WNK4 levels, and suppression of SPAK/OxSR1 expression and phosphorylation. In contrast, in females, there was decreased NCC activity associated with suppression of SPAK/OxSR1 expression and phosphorylation. In hypertensive DSS rats, the ability of females to suppress NCC (in opposition to males) via a SPAK/OxSR1 mechanism likely contributes to their lower magnitude of salt-sensitive hypertension. Collectively, our findings support the existence of sex differences in male versus female rats with NCC regulation during dietary salt intake involving suppression of WNK4 expression in male rats only and the involvement of SPAK/OxSR1 signaling in both males and females.NEW & NOTEWORTHY NCC regulation is sex dependent. In normotensive male and female Sprague-Dawley and Dahl salt-resistant rats, which exhibit dietary Na+-evoked NCC suppression, male rats exhibit decreased WNK4 expression and decreased SPAK and OxSR1 levels, whereas female rats only suppress SPAK and OxSR1. In hypertensive Dahl salt-sensitive rats, the ability of females to suppress NCC (in opposition to males) via a SPAK/OxSR1 mechanism likely contributes to their lower magnitude of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Kiyoung Kim
- Department of Pharmacology and Experimental Therapeutics, Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Kayla M Nist
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Franco Puleo
- Department of Pharmacology and Experimental Therapeutics, Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - James McKenna
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Richard D Wainford
- Department of Pharmacology and Experimental Therapeutics, Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
7
|
Lénárt N, Cserép C, Császár E, Pósfai B, Dénes Á. Microglia-neuron-vascular interactions in ischemia. Glia 2024; 72:833-856. [PMID: 37964690 DOI: 10.1002/glia.24487] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
Cerebral ischemia is a devastating condition that results in impaired blood flow in the brain leading to acute brain injury. As the most common form of stroke, occlusion of cerebral arteries leads to a characteristic sequence of pathophysiological changes in the brain tissue. The mechanisms involved, and comorbidities that determine outcome after an ischemic event appear to be highly heterogeneous. On their own, the processes leading to neuronal injury in the absence of sufficient blood supply to meet the metabolic demand of the cells are complex and manifest at different temporal and spatial scales. While the contribution of non-neuronal cells to stroke pathophysiology is increasingly recognized, recent data show that microglia, the main immune cells of the central nervous system parenchyma, play previously unrecognized roles in basic physiological processes beyond their inflammatory functions, which markedly change during ischemic conditions. In this review, we aim to discuss some of the known microglia-neuron-vascular interactions assumed to contribute to the acute and delayed pathologies after cerebral ischemia. Because the mechanisms of neuronal injury have been extensively discussed in several excellent previous reviews, here we focus on some recently explored pathways that may directly or indirectly shape neuronal injury through microglia-related actions. These discoveries suggest that modulating gliovascular processes in different forms of stroke and other neurological disorders might have presently unexplored therapeutic potential in combination with neuroprotective and flow restoration strategies.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Császár
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
8
|
Althammer F, Roy RK, Kirchner MK, McGrath S, Lira EC, Stern JE. Angiotensin-II drives changes in microglia-vascular interactions in rats with heart failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573045. [PMID: 38187537 PMCID: PMC10769361 DOI: 10.1101/2023.12.22.573045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activation of microglia, the resident immune cells of the central nervous system, leading to the subsequent release of pro-inflammatory cytokines, has been linked to cardiac remodeling, autonomic disbalance, and cognitive deficits in heart failure (HF). While previous studies emphasized the role of hippocampal Angiotensin II (AngII) signaling in HF-induced microglial activation, unanswered mechanistic questions persist. Evidence suggests significant interactions between microglia and local microvasculature, potentially affecting blood-brain barrier integrity and cerebral blood flow regulation. Still, whether the microglial-vascular interface is affected in the brain during HF remains unknow. Using a well-established ischemic HF rat model, we demonstrate increased vessel-associated microglia (VAM) in HF rat hippocampi, which showed heightened expression of AngII AT1a receptors. Acute AngII administration to sham rats induced microglia recruitment to the perivascular space, along with increased expression of TNFa. Conversely, administering an AT1aR blocker to HF rats prevented the recruitment of microglia to the perivascular space, normalizing their levels to those in healthy rats. These results highlight the critical importance of a rather understudied phenomenon (i.e., microglia-vascular interactions in the brain) in the context of the pathophysiology of a highly prevalent cardiovascular disease, and unveil novel potential therapeutic avenues aimed at mitigating neuroinflammation in cardiovascular diseases.
Collapse
|
9
|
Martins Sá RW, Theparambil SM, Dos Santos KM, Christie IN, Marina N, Cardoso BV, Hosford PS, Antunes VR. Salt-loading promotes extracellular ATP release mediated by glial cells in the hypothalamic paraventricular nucleus of rats. Mol Cell Neurosci 2023; 124:103806. [PMID: 36592801 DOI: 10.1016/j.mcn.2022.103806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
Previously, we have shown that purinergic signalling is involved in the control of hyperosmotic-induced sympathoexcitation at the level of the PVN, via activation of P2X receptors. However, the source(s) of ATP that drives osmotically-induced increases in sympathetic outflow remained undetermined. Here, we tested the two competing hypotheses that either (1) higher extracellular ATP in PVN during salt loading (SL) is a result of a failure of ectonucleotidases to metabolize ATP; and/or (2) SL can stimulate PVN astrocytes to release ATP. Rats were salt loaded with a 2 % NaCl solution replacing drinking water up to 4 days, an experimental model known to cause a gradual increase in blood pressure and plasma osmolarity. Immunohistochemical assessment of glial-fibrillary acidic protein (GFAP) revealed increased glial cell reactivity in the PVN of rats after 4 days of high salt exposure. ATP and adenosine release measurements via biosensors in hypothalamic slices showed that baseline ATP release was increased 17-fold in the PVN while adenosine remained unchanged. Disruption of Ca2+-dependent vesicular release mechanisms in PVN astrocytes by virally-driven expression of a dominant-negative SNARE protein decreased the release of ATP. The activity of ectonucleotidases quantified in vitro by production of adenosine from ATP was increased in SL group. Our results showed that SL stimulates the release of ATP in the PVN, at least in part, from glial cells by a vesicle-mediated route and likely contributes to the neural control of circulation during osmotic challenges.
Collapse
Affiliation(s)
- Renato W Martins Sá
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Shefeeq M Theparambil
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Karoline Martins Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Isabel N Christie
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Barbara V Cardoso
- Department of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Patrick S Hosford
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Vagner R Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
10
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
11
|
Freitas FEDA, Batista MAC, Braga DCDA, de Oliveira LB, Antunes VR, Cardoso LM. The gut-brain axis and sodium appetite: Can inflammation-related signaling influence the control of sodium intake? Appetite 2022; 175:106050. [PMID: 35447164 DOI: 10.1016/j.appet.2022.106050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
Sodium is the main cation present in the extracellular fluid. Sodium and water content in the body are responsible for volume and osmotic homeostasis through mechanisms involving sodium and water excretion and intake. When body sodium content decreases below the homeostatic threshold, a condition termed sodium deficiency, highly motivated sodium seeking, and intake occurs. This is termed sodium appetite. Classically, sodium and water intakes are controlled by a number of neuroendocrine mechanisms that include signaling molecules from the renin-angiotensin-aldosterone system acting in the central nervous system (CNS). However, recent findings have shown that sodium and water intakes can also be influenced by inflammatory agents and mediators acting in the CNS. For instance, central infusion of IL-1β or TNF-α can directly affect sodium and water consumption in animal models. Some dietary conditions, such as high salt intake, have been shown to change the intestinal microbiome composition, stimulating the immune branch of the gut-brain axis through the production of inflammatory cytokines, such as IL-17, which can stimulate the brain immune system. In this review, we address the latest findings supporting the hypothesis that immune signaling in the brain could produce a reduction in thirst and sodium appetite and, therefore, contribute to sodium intake control.
Collapse
Affiliation(s)
| | | | | | | | - Vagner Roberto Antunes
- Dept. of Physiology and Biophysics - ICB, University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
12
|
Amraei R, Moreira JD, Wainford RD. Central Gαi 2 Protein Mediated Neuro-Hormonal Control of Blood Pressure and Salt Sensitivity. Front Endocrinol (Lausanne) 2022; 13:895466. [PMID: 35837296 PMCID: PMC9275552 DOI: 10.3389/fendo.2022.895466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Hypertension, a major public health issue, is estimated to contribute to 10% of all deaths worldwide. Further, the salt sensitivity of blood pressure is a critical risk factor for the development of hypertension. The hypothalamic paraventricular nucleus (PVN) coordinates neuro-hormonal responses to alterations in plasma sodium and osmolality and multiple G Protein-Coupled Receptors (GPCRs) are involved in fluid and electrolyte homeostasis. In acute animal studies, our laboratory has shown that central Gαi/o subunit protein signal transduction mediates hypotensive and bradycardic responses and that Gz/q, proteins mediate the release of arginine vasopressin (AVP) and subsequent aquaretic responses to acute pharmacological stimuli. Extending these studies, our laboratory has shown that central Gαi2 proteins selectively mediate the hypotensive, sympathoinhibitory and natriuretic responses to acute pharmacological activation of GPCRs and in response to acute physiological challenges to fluid and electrolyte balance. In addition, following chronically elevated dietary sodium intake, salt resistant rats demonstrate site-specific and subunit-specific upregulation of Gαi2 proteins in the PVN, resulting in sympathoinhibition and normotension. In contrast, chronic dietary sodium intake in salt sensitive animals, which fail to upregulate PVN Gαi2 proteins, results in the absence of dietary sodium-evoked sympathoinhibition and salt sensitive hypertension. Using in situ hybridization, we observed that Gαi2 expressing neurons in parvocellular division of the PVN strongly (85%) colocalize with GABAergic neurons. Our data suggest that central Gαi2 protein-dependent responses to an acute isotonic volume expansion (VE) and elevated dietary sodium intake are mediated by the peripheral sensory afferent renal nerves and do not depend on the anteroventral third ventricle (AV3V) sodium sensitive region or the actions of central angiotensin II type 1 receptors. Our translational human genomic studies have identified three G protein subunit alpha I2 (GNAI2) single nucleotide polymorphisms (SNPs) as potential biomarkers in individuals with salt sensitivity and essential hypertension. Collectively, PVN Gαi2 proteins-gated pathways appear to be highly conserved in salt resistance to counter the effects of acute and chronic challenges to fluid and electrolyte homeostasis on blood pressure via a renal sympathetic nerve-dependent mechanism.
Collapse
Affiliation(s)
- Razie Amraei
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Jesse D. Moreira
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Richard D. Wainford
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
13
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
14
|
Frame AA, Nist KM, Kim K, Kuwabara JT, Wainford RD. Natriuresis During an Acute Intravenous Sodium Chloride Infusion in Conscious Sprague Dawley Rats Is Mediated by a Blood Pressure-Independent α1-Adrenoceptor-Mediated Mechanism. Front Physiol 2022; 12:784957. [PMID: 35111076 PMCID: PMC8802910 DOI: 10.3389/fphys.2021.784957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
The mechanisms that sense alterations in total body sodium content to facilitate sodium homeostasis in response to an acute sodium challenge that does not increase blood pressure have not been fully elucidated. We hypothesized that the renal sympathetic nerves are critical to mediate natriuresis via α1- or β-adrenoceptors signal transduction pathways to maintain sodium balance in the face of acute increases in total body sodium content that do not activate the pressure-natriuresis mechanism. To address this hypothesis, we used acute bilateral renal denervation (RDNX), an anteroventral third ventricle (AV3V) lesion and α1- or β-antagonism during an acute 1M NaCl sodium challenge in conscious male Sprague Dawley rats. An acute 1M NaCl infusion did not alter blood pressure and evoked profound natriuresis and sympathoinhibition. Acute bilateral RDNX attenuated the natriuretic and sympathoinhibitory responses evoked by a 1M NaCl infusion [peak natriuresis (μeq/min) sham 14.5 ± 1.3 vs. acute RDNX: 9.2 ± 1.4, p < 0.05; plasma NE (nmol/L) sham control: 44 ± 4 vs. sham 1M NaCl infusion 11 ± 2, p < 0.05; acute RDNX control: 42 ± 6 vs. acute RDNX 1M NaCl infusion 25 ± 3, p < 0.05]. In contrast, an AV3V lesion did not impact the cardiovascular, renal excretory or sympathoinhibitory responses to an acute 1M NaCl infusion. Acute i.v. α1-adrenoceptor antagonism with terazosin evoked a significant drop in baseline blood pressure and significantly attenuated the natriuretic response to a 1M NaCl load [peak natriuresis (μeq/min) saline 17.2 ± 1.4 vs. i.v. terazosin 7.8 ± 2.5, p < 0.05]. In contrast, acute β-adrenoceptor antagonism with i.v. propranolol infusion did not impact the cardiovascular or renal excretory responses to an acute 1M NaCl infusion. Critically, the natriuretic response to an acute 1M NaCl infusion was significantly blunted in rats receiving a s.c. infusion of the α1-adrenoceptor antagonist terazosin at a dose that did not lower baseline blood pressure [peak natriuresis (μeq/min) sc saline: 18 ± 1 vs. sc terazosin 7 ± 2, p < 0.05]. Additionally, a s.c. infusion of the α1-adrenoceptor antagonist terazosin further attenuated the natriuretic response to a 1M NaCl infusion in acutely RDNX animals. Collectively these data indicate a specific role of a blood pressure-independent renal sympathetic nerve-dependent α1-adrenoceptor-mediated pathway in the natriuretic and sympathoinhibitory responses evoked by acute increases in total body sodium.
Collapse
Affiliation(s)
- Alissa A. Frame
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Kayla M. Nist
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Kiyoung Kim
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Jill T. Kuwabara
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Richard D. Wainford
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
- *Correspondence: Richard D. Wainford,
| |
Collapse
|
15
|
Moreira JD, Nist KM, Carmichael CY, Kuwabara JT, Wainford RD. Sensory Afferent Renal Nerve Activated Gαi 2 Subunit Proteins Mediate the Natriuretic, Sympathoinhibitory and Normotensive Responses to Peripheral Sodium Challenges. Front Physiol 2021; 12:771167. [PMID: 34916958 PMCID: PMC8669768 DOI: 10.3389/fphys.2021.771167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/09/2021] [Indexed: 11/15/2022] Open
Abstract
We have previously reported that brain Gαi2 subunit proteins are required to maintain sodium homeostasis and are endogenously upregulated in the hypothalamic paraventricular nucleus (PVN) in response to increased dietary salt intake to maintain a salt resistant phenotype in rats. However, the origin of the signal that drives the endogenous activation and up-regulation of PVN Gαi2 subunit protein signal transduction pathways is unknown. By central oligodeoxynucleotide (ODN) administration we show that the pressor responses to central acute administration and central infusion of sodium chloride occur independently of brain Gαi2 protein pathways. In response to an acute volume expansion, we demonstrate, via the use of selective afferent renal denervation (ADNX) and anteroventral third ventricle (AV3V) lesions, that the sensory afferent renal nerves, but not the sodium sensitive AV3V region, are mechanistically involved in Gαi2 protein mediated natriuresis to an acute volume expansion [peak natriuresis (μeq/min) sham AV3V: 43 ± 4 vs. AV3V 45 ± 4 vs. AV3V + Gαi2 ODN 25 ± 4, p < 0.05; sham ADNX: 43 ± 4 vs. ADNX 23 ± 6, AV3V + Gαi2 ODN 25 ± 3, p < 0.05]. Furthermore, in response to chronically elevated dietary sodium intake, endogenous up-regulation of PVN specific Gαi2 proteins does not involve the AV3V region and is mediated by the sensory afferent renal nerves to counter the development of the salt sensitivity of blood pressure (MAP [mmHg] 4% NaCl; Sham ADNX 124 ± 4 vs. ADNX 145 ± 4, p < 0.05; Sham AV3V 125 ± 4 vs. AV3V 121 ± 5). Additionally, the development of the salt sensitivity of blood pressure following central ODN-mediated Gαi2 protein down-regulation occurs independently of the actions of the brain angiotensin II type 1 receptor. Collectively, our data suggest that in response to alterations in whole body sodium the peripheral sensory afferent renal nerves, but not the central AV3V sodium sensitive region, evoke the up-regulation and activation of PVN Gαi2 protein gated pathways to maintain a salt resistant phenotype. As such, both the sensory afferent renal nerves and PVN Gαi2 protein gated pathways, represent potential targets for the treatment of the salt sensitivity of blood pressure.
Collapse
Affiliation(s)
- Jesse D. Moreira
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Medicine, School of Medicine, Boston University, Boston, MA, United States
| | - Kayla M. Nist
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Anatomy & Neurobiology, School of Medicine, Boston University, Boston, MA, United States
| | - Casey Y. Carmichael
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| | - Jill T. Kuwabara
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| | - Richard D. Wainford
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
16
|
Cao Y, Yu Y, Xue B, Wang Y, Chen X, Beltz TG, Johnson AK, Wei SG. IL (Interleukin)-17A Acts in the Brain to Drive Neuroinflammation, Sympathetic Activation, and Hypertension. Hypertension 2021; 78:1450-1462. [PMID: 34628936 DOI: 10.1161/hypertensionaha.121.18219] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yiling Cao
- Department of Internal Medicine (Y.C., Y.Y., S.-G.W.), University of Iowa Carver College of Medicine
| | - Yang Yu
- Department of Internal Medicine (Y.C., Y.Y., S.-G.W.), University of Iowa Carver College of Medicine
| | - Baojian Xue
- Psychological and Brain Sciences (B.X., T.G.B., A.K.J.), University of Iowa Carver College of Medicine
| | - Ye Wang
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University, China (Y.W.)
| | - Xiaolei Chen
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China (X.C.)
| | - Terry G Beltz
- Psychological and Brain Sciences (B.X., T.G.B., A.K.J.), University of Iowa Carver College of Medicine
| | - Alan Kim Johnson
- Psychological and Brain Sciences (B.X., T.G.B., A.K.J.), University of Iowa Carver College of Medicine.,Abboud Cardiovascular Research Center (A.K.J., S.-G.W.), University of Iowa Carver College of Medicine.,Iowa Neuroscience Institute (A.K.J., S.-G.W.), University of Iowa Carver College of Medicine
| | - Shun-Guang Wei
- Department of Internal Medicine (Y.C., Y.Y., S.-G.W.), University of Iowa Carver College of Medicine.,Abboud Cardiovascular Research Center (A.K.J., S.-G.W.), University of Iowa Carver College of Medicine.,Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University, China (Y.W.)
| |
Collapse
|
17
|
Gao HL, Yu XJ, Hu HB, Yang QW, Liu KL, Chen YM, Zhang Y, Zhang DD, Tian H, Zhu GQ, Qi J, Kang YM. Apigenin Improves Hypertension and Cardiac Hypertrophy Through Modulating NADPH Oxidase-Dependent ROS Generation and Cytokines in Hypothalamic Paraventricular Nucleus. Cardiovasc Toxicol 2021; 21:721-736. [PMID: 34076830 DOI: 10.1007/s12012-021-09662-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/17/2021] [Indexed: 12/26/2022]
Abstract
Apigenin, identified as 4', 5, 7-trihydroxyflavone, is a natural flavonoid compound that has many interesting pharmacological activities and nutraceutical potential including anti-inflammatory and antioxidant functions. Chronic, low-grade inflammation and oxidative stress are involved in both the initiation and progression of hypertension and hypertension-induced cardiac hypertrophy. However, whether or not apigenin improves hypertension and cardiac hypertrophy through modulating NADPH oxidase-dependent reactive oxygen species (ROS) generation and inflammation in hypothalamic paraventricular nucleus (PVN) has not been reported. This study aimed to investigate the effects of apigenin on hypertension in spontaneously hypertensive rats (SHRs) and its possible central mechanism of action. SHRs and Wistar-Kyoto (WKY) rats were randomly assigned and treated with bilateral PVN infusion of apigenin or vehicle (artificial cerebrospinal fluid) via osmotic minipumps (20 μg/h) for 4 weeks. The results showed that after PVN infusion of apigenin, the mean arterial pressure (MAP), heart rate, plasma norepinephrine (NE), Beta 1 receptor in kidneys, level of phosphorylation of PKA in the ventricular tissue and cardiac hypertrophy, perivascular fibrosis, heart level of oxidative stress, PVN levels of oxidative stress, interleukin 1β (IL-1β), interleukin 6 (IL-6), iNOS, monocyte chemotactic protein 1 (MCP-1), tyrosine hydroxylase (TH), NOX2 and NOX4 were attenuated and PVN levels of interleukin 10 (IL-10), superoxide dismutase 1 (Cu/Zn-SOD) and the 67-kDa isoform of glutamate decarboxylase (GAD67) were increased. These results revealed that apigenin improves hypertension and cardiac hypertrophy in SHRs which are associated with the down-regulation of NADPH oxidase-dependent ROS generation and inflammation in the PVN.
Collapse
Affiliation(s)
- Hong-Li Gao
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Xiao-Jing Yu
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Han-Bo Hu
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Qian-Wen Yang
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Kai-Li Liu
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Yan-Mei Chen
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Yan Zhang
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Dong-Dong Zhang
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Hua Tian
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China
| | - Guo-Qing Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Jie Qi
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China.
| | - Yu-Ming Kang
- Key Laboratory of Environment and Genes Related To Diseases of Education Ministry of China, Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Xi'an, 710061, China.
| |
Collapse
|
18
|
Gao H, Bigalke J, Jiang E, Fan Y, Chen B, Chen QH, Shan Z. TNFα Triggers an Augmented Inflammatory Response in Brain Neurons from Dahl Salt-Sensitive Rats Compared with Normal Sprague Dawley Rats. Cell Mol Neurobiol 2021; 42:1787-1800. [PMID: 33625627 PMCID: PMC8382783 DOI: 10.1007/s10571-021-01056-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/04/2021] [Indexed: 12/23/2022]
Abstract
Tumor Necrosis Factor (TNF)-α is a proinflammatory cytokine (PIC) and has been implicated in a variety of illness including cardiovascular disease. The current study investigated the inflammatory response trigged by TNFα in both cultured brain neurons and the hypothalamic paraventricular nucleus (PVN), a key cardiovascular relevant brain area, of the Sprague Dawley (SD) rats. Our results demonstrated that TNFα treatment induces a dose- and time-dependent increase in mRNA expression of PICs including Interleukin (IL)-1β and Interleukin-6 (IL6); chemokines including C-C Motif Chemokine Ligand 5 (CCL5) and C-C Motif Chemokine Ligand 12 (CCL12), inducible nitric oxide synthase (iNOS), as well as transcription factor NF-kB in cultured brain neurons from neonatal SD rats. Consistent with this finding, immunostaining shows that TNFα treatment increases immunoreactivity of IL1β, CCL5, iNOS and stimulates activation or expression of NF-kB, in both cultured brain neurons and the PVN of adult SD rats. We further compared mRNA expression of the aforementioned genes in basal level as well as in response to TNFα challenge between SD rats and Dahl Salt-sensitive (Dahl-S) rats, an animal model of salt-sensitive hypertension. Dahl-S brain neurons presented higher baseline levels as well as greater response to TNFα challenge in mRNA expression of CCL5, iNOS and IL1β. Furthermore, central administration of TNFα caused significant higher response in CCL12 in the PVN of Dahl-S rats. The increased inflammatory response to TNFα in Dahl-S rats may be indicative of an underlying mechanism for enhanced pressor reactivity to salt intake in the Dahl-S rat model.
Collapse
Affiliation(s)
- Huanjia Gao
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Jeremy Bigalke
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA
| | - Enshe Jiang
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,Institute of Nursing and Health, Henan University, Henan, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, Henan University, Henan, China
| | - Yuanyuan Fan
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,School of Life Sciences, Henan University, Henan, China
| | - Bojun Chen
- Department of Emergency, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing-Hui Chen
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA.,Health Research Institute, Michigan Technological University, Houghton, MI, 49931, USA
| | - Zhiying Shan
- Department of Kinesiology & Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA. .,Health Research Institute, Michigan Technological University, Houghton, MI, 49931, USA.
| |
Collapse
|
19
|
Ahmari N, Hayward LF, Zubcevic J. The importance of bone marrow and the immune system in driving increases in blood pressure and sympathetic nerve activity in hypertension. Exp Physiol 2020; 105:1815-1826. [PMID: 32964557 DOI: 10.1113/ep088247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/01/2020] [Indexed: 12/27/2022]
Abstract
NEW FINDINGS What is the topic of this review? This manuscript provides a review of the current understanding of the role of the sympathetic nervous system in regulation of bone marrow-derived immune cells and the effect that the infiltrating bone marrow cells may have on perpetuation of the sympathetic over-activation in hypertension. What advances does it highlight? We highlight the recent advances in understanding of the neuroimmune interactions both peripherally and centrally as they relate to blood pressure control. ABSTRACT The sympathetic nervous system (SNS) plays a crucial role in maintaining physiological homeostasis, in part by regulating, integrating and orchestrating processes between many physiological systems, including the immune system. Sympathetic nerves innervate all primary and secondary immune organs, and all cells of the immune system express β-adrenoreceptors. In turn, immune cells can produce cytokines, chemokines and neurotransmitters capable of modulating neuronal activity and, ultimately, SNS activity. Thus, the essential role of the SNS in the regulation of innate and adaptive immune functions is mediated, in part, via β-adrenoreceptor-induced activation of bone marrow cells by noradrenaline. Interestingly, both central and systemic inflammation are well-established hallmarks of hypertension and its co-morbidities, including an inflammatory process involving the transmigration and infiltration of immune cells into tissues. We propose that physiological states that prolong β-adrenoreceptor activation in bone marrow can disrupt neuroimmune homeostasis and impair communication between the immune system and SNS, leading to immune dysregulation, which, in turn, is sustained via a central mechanism involving neuroinflammation.
Collapse
Affiliation(s)
- Niousha Ahmari
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Linda F Hayward
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA.,Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Jasenka Zubcevic
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA.,Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
20
|
Salt-dependent hypertension and inflammation: targeting the gut-brain axis and the immune system with Brazilian green propolis. Inflammopharmacology 2020; 28:1163-1182. [PMID: 32785827 PMCID: PMC8826348 DOI: 10.1007/s10787-020-00742-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/30/2020] [Indexed: 01/22/2023]
Abstract
Systemic arterial hypertension (SAH) is a major health problem around the world and its development has been associated with exceeding salt consumption by the modern society. The mechanisms by which salt consumption increase blood pressure (BP) involve several homeostatic systems but many details have not yet been fully elucidated. Evidences accumulated over the last 60 decades raised the involvement of the immune system in the hypertension development and opened a range of possibilities for new therapeutic targets. Green propolis is a promising natural product with potent anti-inflammatory properties acting on specific targets, most of them participating in the gut-brain axis of the sodium-dependent hypertension. New anti-hypertensive products reinforce the therapeutic arsenal improving the corollary of choices, especially in those cases where patients are resistant or refractory to conventional therapy. This review sought to bring the newest advances in the field articulating evidences that show a cross-talking between inflammation and the central mechanisms involved with the sodium-dependent hypertension as well as the stablished actions of green propolis and some of its biologically active compounds on the immune cells and cytokines that would be involved with its anti-hypertensive properties.
Collapse
|
21
|
Carmichael CY, Kuwabara JT, Pascale CL, Moreira JD, Mahne SE, Kapusta DR, Rosene DL, Williams JS, Cunningham JT, Wainford RD. Hypothalamic Paraventricular Nucleus Gαi 2 (Guanine Nucleotide-Binding Protein Alpha Inhibiting Activity Polypeptide 2) Protein-Mediated Neural Control of the Kidney and the Salt Sensitivity of Blood Pressure. Hypertension 2020; 75:1002-1011. [PMID: 32148128 PMCID: PMC7329357 DOI: 10.1161/hypertensionaha.119.13777] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We have previously reported that in salt-resistant rat phenotypes brain, Gαi2 (guanine nucleotide-binding protein alpha inhibiting activity polypeptide 2) proteins are required to maintain blood pressure and sodium balance. However, the impact of hypothalamic paraventricular nucleus (PVN) Gαi2 proteins on the salt sensitivity of blood pressure is unknown. Here, by the bilateral PVN administration of a targeted Gαi2 oligodeoxynucleotide, we show that PVN-specific Gαi2 proteins are required to facilitate the full natriuretic response to an acute volume expansion (peak natriuresis [μeq/min] scrambled (SCR) oligodeoxynucleotide 41±3 versus Gαi2 oligodeoxynucleotide 18±4; P<0.05) via a renal nerve-dependent mechanism. Furthermore, in response to chronically elevated dietary sodium intake, PVN-specific Gαi2 proteins are essential to counter renal nerve-dependent salt-sensitive hypertension (mean arterial pressure [mm Hg] 8% NaCl; SCR oligodeoxynucleotide 128±2 versus Gαi2 oligodeoxynucleotide 147±3; P<0.05). This protective pathway involves activation of PVN Gαi2 signaling pathways, which mediate sympathoinhibition to the blood vessels and kidneys (renal norepinephrine [pg/mg] 8% NaCl; SCR oligodeoxynucleotide 375±39 versus Gαi2 oligodeoxynucleotide 850±27; P<0.05) and suppression of the activity of the sodium chloride cotransporter assessed as peak natriuresis to hydrochlorothiazide. Additionally, central oligodeoxynucleotide-mediated Gαi2 protein downregulation prevented PVN parvocellular neuron activation, assessed by FosB immunohistochemistry, in response to increased dietary salt intake. In our analysis of the UK BioBank data set, it was observed that 2 GNAI2 single nucleotide polymorphism (SNP) (rs2298952, P=0.041; rs4547694, P=0.017) significantly correlate with essential hypertension. Collectively, our data suggest that selective targeting and activation of PVN Gαi2 proteins is a novel therapeutic approach for the treatment of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Casey Y Carmichael
- The Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Jill T Kuwabara
- The Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Crissey L Pascale
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Jesse D Moreira
- The Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
- Department of Health Sciences, Boston University Sargent College, Boston, Massachusetts
| | - Sarah E Mahne
- The Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Daniel R Kapusta
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes, Hypertension, Brigham and Women’s, Harvard Medical School, Boston, Massachusetts
| | - J. Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Richard D Wainford
- The Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
22
|
Moreira JD, Chaudhary P, Frame AA, Puleo F, Nist KM, Abkin EA, Moore TL, George JC, Wainford RD. Inhibition of microglial activation in rats attenuates paraventricular nucleus inflammation in Gαi 2 protein-dependent, salt-sensitive hypertension. Exp Physiol 2019; 104:1892-1910. [PMID: 31631436 PMCID: PMC6884700 DOI: 10.1113/ep087924] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS • What is the central question of this study? We hypothesized that central inflammatory processes that involve activation of microglia and astrocytes contribute to the development of Gαi2 protein-dependent, salt-sensitive hypertension. • What is the main finding and its importance? The main finding is that PVN-specific inflammatory processes, driven by microglial activation, appear to be linked to the development of Gαi2 protein-dependent, salt-sensitive hypertension in Sprague-Dawley rats. This finding might reveal new mechanistic targets in the treatment of hypertension. ABSTRACT The central mechanisms underlying salt-sensitive hypertension, a significant public health issue, remain to be established. Researchers in our laboratory have reported that hypothalamic paraventricular nucleus (PVN) Gαi2 proteins mediate the sympathoinhibitory and normotensive responses to high sodium intake in salt-resistant rats. Given the recent evidence of central inflammation in animal models of hypertension, we hypothesized that PVN inflammation contributes to Gαi2 protein-dependent, salt-sensitive hypertension. Male Sprague-Dawley rats received chronic intracerebroventricular infusions of a targeted Gαi2 or control scrambled oligodeoxynucleotide (ODN) and were maintained for 7 days on a normal-salt (NS; 0.6% NaCl) or high-salt (HS; 4% NaCl) diet; in subgroups on HS, intracerebroventricular minocycline (microglial inhibitor) was co-infused with ODNs. Radiotelemetry was used in subgroups of rats to measure mean arterial pressure (MAP) chronically. In a separate group of rats, plasma noradrenaline, plasma renin activity, urinary angiotensinogen and mRNA levels of the PVN pro-inflammatory cytokines TNFα, IL-1β and IL-6 and the anti-inflammatory cytokine IL-10 were assessed. In additional groups, immunohistochemistry was performed for markers of PVN and subfornical organ microglial activation and cytokine levels and PVN astrocyte activation. High salt intake evoked salt-sensitive hypertension, increased plasma noradrenaline, PVN pro-inflammatory cytokine mRNA upregulation, anti-inflammatory cytokine mRNA downregulation and PVN-specific microglial activation in rats receiving a targeted Gαi2 but not scrambled ODN. Minocycline co-infusion significantly attenuated the increase in MAP and abolished the increase in plasma noradrenaline and inflammation in Gαi2 ODN-infused animals on HS. Our data suggest that central Gαi2 protein prevents microglial-mediated PVN inflammation and the development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Jesse D. Moreira
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Health SciencesBoston University Sargent CollegeBostonMAUSA
| | - Parul Chaudhary
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Alissa A. Frame
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Franco Puleo
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Kayla M. Nist
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMAUSA
| | - Eric A. Abkin
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Health SciencesBoston University Sargent CollegeBostonMAUSA
| | - Tara L. Moore
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMAUSA
| | - Jonique C. George
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Richard D. Wainford
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Health SciencesBoston University Sargent CollegeBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| |
Collapse
|