101
|
Hurley SW, Beltz TG, Guo F, Xue B, Johnson AK. Amphetamine-induced sensitization of hypertension and lamina terminalis neuroinflammation. Am J Physiol Regul Integr Comp Physiol 2020; 318:R649-R656. [PMID: 32048863 DOI: 10.1152/ajpregu.00233.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Psychomotor stimulants are prescribed for many medical conditions, including obesity, sleep disorders, and attention-deficit/hyperactivity disorder. However, despite their acknowledged therapeutic utility, these stimulants are frequently abused, and their use can have both short- and long-term negative consequences. Although stimulants such as amphetamines acutely elevate blood pressure, it is unclear whether they cause any long-term effects on cardiovascular function after use has been discontinued. Previous work in our laboratory has demonstrated that physiological and psychosocial stressors will produce sensitization of the hypertensive response, a heightened pressor response to a hypertensinogenic stimulus delivered after stressor exposure. Here, we tested whether pretreatment with amphetamine for 1 wk can sensitize the hypertensive response in rats. We found that repeated amphetamine administration induced and maintained sensitization of the pressor response to angiotensin II following a 7-day delay after amphetamine injections were terminated. We also found that amphetamine pretreatment altered mRNA expression for molecular markers associated with neuroinflammation and renin-angiotensin-aldosterone system (RAAS) activation in the lamina terminalis, a brain region implicated in the control of sympathetic nervous system tone and blood pressure. The results indicated amphetamine upregulated mRNA expression underlying neuroinflammation and, to a lesser degree, message for components of the RAAS in the lamina terminalis. However, we found no changes in mRNA expression in the paraventricular nucleus. These results suggest that a history of stimulant use may predispose individuals to developing hypertension by promoting neuroinflammation and upregulating activity of the RAAS in the lamina terminalis.
Collapse
Affiliation(s)
- Seth W Hurley
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, North Carolina
| | - Terry G Beltz
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Fang Guo
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa.,Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa.,Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa.,The François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, Iowa
| |
Collapse
|
102
|
Abstract
Hypertension is an important risk factor for cardiovascular morbidity and mortality and for events such as myocardial infarction, stroke, heart failure and chronic kidney disease and is a major determinant of disability-adjusted life-years. Despite the importance of hypertension, the pathogenesis of essential hypertension, which involves the complex interaction of several mechanisms, is still poorly understood. Evidence suggests that interplay between bone marrow, microglia and immune mediators underlies the development of arterial hypertension, in particular through mechanisms involving cytokines and peptides, such as neuropeptide Y, substance P, angiotensin II and angiotensin-(1-7). Chronic psychological stress also seems to have a role in increasing the risk of hypertension, probably through the activation of neuroimmune pathways. In this Review, we summarize the available data on the possible role of neuroimmune crosstalk in the origin and maintenance of arterial hypertension and discuss the implications of this crosstalk for recovery and rehabilitation after cardiac and cerebral injuries.
Collapse
|
103
|
Iulita MF, Duchemin S, Vallerand D, Barhoumi T, Alvarez F, Istomine R, Laurent C, Youwakim J, Paradis P, Arbour N, Piccirillo CA, Schiffrin EL, Girouard H. CD4 + Regulatory T Lymphocytes Prevent Impaired Cerebral Blood Flow in Angiotensin II-Induced Hypertension. J Am Heart Assoc 2020; 8:e009372. [PMID: 30572753 PMCID: PMC6405729 DOI: 10.1161/jaha.118.009372] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Immune cells are key regulators of the vascular inflammatory response characteristic of hypertension. In hypertensive rodents, regulatory T lymphocytes (Treg, CD4+CD25+) prevented vascular injury, cardiac damage, and endothelial dysfunction of mesenteric arteries. Whether Treg modulate the cerebrovascular damage induced by hypertension is unknown. Methods and Results C57BL/6 mice were perfused with angiotensin II (Ang II; 1000 ng/kg per minute) for 14 days and adoptive transfer of 3×105CD4+CD25+ T cells was performed via 2 intravenous injections. Control mice received a sham surgery and PBS. Treg prevented Ang II‐induced neurovascular uncoupling (P<0.05) and endothelial impairment (P<0.05), evaluated by laser Doppler flowmetry in the somatosensory cortex. The neuroprotective effect of Treg was abolished when they were isolated from mice deficient in interleukin‐10. Administration of interleukin‐10 (60 ng/d) to hypertensive mice prevented Ang II‐induced neurovascular uncoupling (P<0.05). Treg adoptive transfer also diminished systemic inflammation induced by Ang II (P<0.05), examined with a peripheral blood cytokine array. Mice receiving Ang II + Treg exhibited reduced numbers of Iba‐1+ cells in the brain cortex (P<0.05) and hippocampus (P<0.001) compared with mice infused only with Ang II. Treg prevented the increase in cerebral superoxide radicals. Overall, these effects did not appear to be directly modulated by Treg accumulating in the brain parenchyma, because only a nonsignificant number of Treg were detected in brain. Instead, Treg penetrated peripheral tissues such as the kidney, inguinal lymph nodes, and the spleen. Conclusions Treg prevent impaired cerebrovascular responses in Ang II‐induced hypertension. The neuroprotective effects of Treg involve the modulation of inflammation in the brain and periphery.
Collapse
Affiliation(s)
- M Florencia Iulita
- 1 Department of Neurosciences Université de Montréal Montréal Canada.,2 Groupe de recherche sur le système nerveux central (GRSNC) Université de Montréal Montréal Canada
| | - Sonia Duchemin
- 4 Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| | - Diane Vallerand
- 4 Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| | - Tlili Barhoumi
- 5 Lady Davis Institute for Medical Research McGill University Montréal Canada
| | - Fernando Alvarez
- 6 Centre of Excellence in Translational Immunology Research Institute of McGill University Health Centre McGill University Montréal Canada.,7 Department of Microbiology and Immunology McGill University Montréal Canada
| | - Roman Istomine
- 6 Centre of Excellence in Translational Immunology Research Institute of McGill University Health Centre McGill University Montréal Canada.,7 Department of Microbiology and Immunology McGill University Montréal Canada
| | - Cyril Laurent
- 1 Department of Neurosciences Université de Montréal Montréal Canada.,3 Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal Canada
| | - Jessica Youwakim
- 4 Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| | - Pierre Paradis
- 5 Lady Davis Institute for Medical Research McGill University Montréal Canada
| | - Nathalie Arbour
- 1 Department of Neurosciences Université de Montréal Montréal Canada.,3 Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal Canada
| | - Ciriaco A Piccirillo
- 6 Centre of Excellence in Translational Immunology Research Institute of McGill University Health Centre McGill University Montréal Canada.,7 Department of Microbiology and Immunology McGill University Montréal Canada
| | - Ernesto L Schiffrin
- 5 Lady Davis Institute for Medical Research McGill University Montréal Canada.,8 Department of Medicine Sir Mortimer B. Davis-Jewish General Hospital McGill University Montréal Canada
| | - Hélène Girouard
- 2 Groupe de recherche sur le système nerveux central (GRSNC) Université de Montréal Montréal Canada.,4 Department of Pharmacology and Physiology Université de Montréal Montréal Canada.,9 Centre de recherche de l'Institut universitaire de gériatrie de Montréal Canada
| |
Collapse
|
104
|
Díaz HS, Toledo C, Andrade DC, Marcus NJ, Del Rio R. Neuroinflammation in heart failure: new insights for an old disease. J Physiol 2020; 598:33-59. [PMID: 31671478 DOI: 10.1113/jp278864] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 08/25/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome affecting roughly 26 million people worldwide. Increased sympathetic drive is a hallmark of HF and is associated with disease progression and higher mortality risk. Several mechanisms contribute to enhanced sympathetic activity in HF, but these pathways are still incompletely understood. Previous work suggests that inflammation and activation of the renin-angiotensin system (RAS) increases sympathetic drive. Importantly, chronic inflammation in several brain regions is commonly observed in aged populations, and a growing body of evidence suggests neuroinflammation plays a crucial role in HF. In animal models of HF, central inhibition of RAS and pro-inflammatory cytokines normalizes sympathetic drive and improves cardiac function. The precise molecular and cellular mechanisms that lead to neuroinflammation and its effect on HF progression remain undetermined. This review summarizes the most recent advances in the field of neuroinflammation and autonomic control in HF. In addition, it focuses on cellular and molecular mediators of neuroinflammation in HF and in particular on brain regions involved in sympathetic control. Finally, we will comment on what is known about neuroinflammation in the context of preserved vs. reduced ejection fraction HF.
Collapse
Affiliation(s)
- Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Noah J Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, USA
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
105
|
Sharma RK, Yang T, Oliveira AC, Lobaton GO, Aquino V, Kim S, Richards EM, Pepine CJ, Sumners C, Raizada MK. Microglial Cells Impact Gut Microbiota and Gut Pathology in Angiotensin II-Induced Hypertension. Circ Res 2019; 124:727-736. [PMID: 30612527 DOI: 10.1161/circresaha.118.313882] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Increased microglial activation and neuroinflammation within autonomic brain regions have been implicated in sustained hypertension, and their inhibition by minocycline-an anti-inflammatory antibiotic-produces beneficial effects. These observations led us to propose a dysfunctional brain-gut communication hypothesis for hypertension. However, it has been difficult to reconcile whether an anti-inflammatory or antimicrobial action is the primary beneficial effect of minocycline in hypertension. Accordingly, we utilized chemically modified tetracycline-3 (CMT-3)-a derivative of tetracycline that has potent anti-inflammatory activity-to address this question. OBJECTIVE Test the hypothesis that central administration of CMT-3 would inhibit microglial activation, attenuate neuroinflammation, alter selective gut microbial communities, protect the gut wall from developing hypertension-associated pathology, and attenuate hypertension. METHODS AND RESULTS Rats were implanted with radiotelemetry devices for recording mean arterial pressure. Ang II (angiotensin II) was infused subcutaneously using osmotic mini-pumps to induce hypertension. Another osmotic mini-pump was surgically implanted to infuse CMT-3 intracerebroventricularly. Intracerebroventricular CMT- 3 infusion was also investigated in SHR (spontaneously hypertensive rats). Physiological, pathological, immunohistological parameters, and fecal microbiota were analyzed. Intracerebroventricular CMT-3 significantly inhibited Ang II-induced increases in number of microglia, their activation, and proinflammatory cytokines in the paraventricular nucleus of hypothalamus. Further, intracerebroventricular CMT-3 attenuated increased mean arterial pressure, normalized sympathetic activity, and left ventricular hypertrophy in Ang II rats, as well as in the SHR. Finally, CMT-3 beneficially restored certain gut microbial communities altered by Ang II and attenuated pathological alterations in gut wall. CONCLUSIONS These observations demonstrate that inhibition of microglial activation alone was sufficient to induce significant antihypertensive effects. This was associated with unique changes in gut microbial communities and profound attenuation of gut pathology. They suggest, for the first time, a link between microglia and certain microbial communities that may have implications for treatment of hypertension.
Collapse
Affiliation(s)
- Ravindra K Sharma
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Tao Yang
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Aline C Oliveira
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Gilberto O Lobaton
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Victor Aquino
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Seungbum Kim
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Elaine M Richards
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Carl J Pepine
- Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville
| | - Colin Sumners
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Mohan K Raizada
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| |
Collapse
|
106
|
Sumners C, Alleyne A, Rodríguez V, Pioquinto DJ, Ludin JA, Kar S, Winder Z, Ortiz Y, Liu M, Krause EG, de Kloet AD. Brain angiotensin type-1 and type-2 receptors: cellular locations under normal and hypertensive conditions. Hypertens Res 2019; 43:281-295. [PMID: 31853042 DOI: 10.1038/s41440-019-0374-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 12/15/2022]
Abstract
Brain angiotensin-II (Ang-II) type-1 receptors (AT1Rs), which exert profound effects on normal cardiovascular, fluid, and metabolic homeostasis, are overactivated in and contribute to chronic sympathoexcitation and hypertension. Accumulating evidence indicates that the activation of Ang-II type-2 receptors (AT2Rs) in the brain exerts effects that are opposite to those of AT1Rs, lowering blood pressure, and reducing hypertension. Thus, it would be interesting to understand the relative cellular localization of AT1R and AT2R in the brain under normal conditions and whether this localization changes during hypertension. Here, we developed a novel AT1aR-tdTomato reporter mouse strain in which the location of brain AT1aR was largely consistent with that determined in the previous studies. This AT1aR-tdTomato reporter mouse strain was crossed with our previously described AT2R-eGFP reporter mouse strain to yield a novel dual AT1aR/AT2R reporter mouse strain, which allowed us to determine that AT1aR and AT2R are primarily localized to different populations of neurons in brain regions controlling cardiovascular, fluid, and metabolic homeostasis. Using the individual AT1aR-tdTomato reporter mice, we also demonstrated that during hypertension induced by the administration of deoxycorticosterone acetate-salt, there was no shift in the expression of AT1aR from neurons to microglia or astrocytes in the paraventricular nucleus, a brain area important for sympathetic regulation. Using AT2R-eGFP reporter mice under similar hypertensive conditions, we demonstrated that the same was true of AT2R expression in the nucleus of the solitary tract (NTS), an area critical for baroreflex control. Collectively, these findings provided a novel means to assess the colocalization of AT1R and AT2R in the brain and a novel view of their cellular localization in hypertension.
Collapse
Affiliation(s)
- Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Amy Alleyne
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Vermalí Rodríguez
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - David J Pioquinto
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Jacob A Ludin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Shormista Kar
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Zachary Winder
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.,Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Yuma Ortiz
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Meng Liu
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
107
|
Leenen FHH, Wang HW, Hamlyn JM. Sodium pumps, ouabain and aldosterone in the brain: A neuromodulatory pathway underlying salt-sensitive hypertension and heart failure. Cell Calcium 2019; 86:102151. [PMID: 31954234 DOI: 10.1016/j.ceca.2019.102151] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/15/2019] [Indexed: 02/08/2023]
Abstract
Accumulating evidence obtained over the last three decades has revealed a neuroendocrine system in the brain that mediates long term increases in blood pressure. The system involves distinct ion transport pathways including the alpha-2 isoform of the Na,K pump and epithelial sodium channels, as well as critical hormone elements such as angiotensin II, aldosterone, mineralocorticoid receptors and endogenous ouabain. Activation of this system either by circulating or central sodium ions and/or angiotensin II leads to a cascading sequence of events that begins in the hypothalamus and involves the participation of several brain nuclei including the subfornical organ, supraoptic and paraventricular nuclei and the rostral ventral medulla. Key events include heightened aldosterone synthesis and mineralocorticoid receptor activation, upregulation of epithelial sodium channels, augmented synthesis and secretion of endogenous ouabain from hypothalamic magnocellular neurons, and sustained increases in sympathetic outflow. The latter step depends upon increased production of angiotensin II and the primary amplification of angiotensin II type I receptor signaling from the paraventricular nucleus to the rostral ventral lateral medulla. The transmission of sympathetic traffic is secondarily amplified in the periphery by increased short- and long-term potentiation in sympathetic ganglia and by sustained actions of endogenous ouabain in the vascular wall that augment expression of sodium calcium exchange, increase cytosolic Ca2+ and heighten myogenic tone and contractility. Upregulation of this multi-amplifier system participates in forms of hypertension where salt, angiotensin and/or aldosterone are elevated and contributes to adverse outcomes in heart failure.
Collapse
Affiliation(s)
- Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - John M Hamlyn
- Department of Physiology, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
108
|
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
|
109
|
Brain perivascular macrophages: connecting inflammation to autonomic activity in hypertension. Hypertens Res 2019; 43:148-150. [PMID: 31745290 DOI: 10.1038/s41440-019-0359-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/17/2022]
|
110
|
Fu MH, Chen IC, Lee CH, Wu CW, Lee YC, Kung YC, Hung CY, Wu KLH. Anti-neuroinflammation ameliorates systemic inflammation-induced mitochondrial DNA impairment in the nucleus of the solitary tract and cardiovascular reflex dysfunction. J Neuroinflammation 2019; 16:224. [PMID: 31729994 PMCID: PMC6858639 DOI: 10.1186/s12974-019-1623-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/24/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Decreased heart rate variability (HRV) leads to cardiovascular diseases and increased mortality in clinical studies. However, the underlying mechanisms are still inconclusive. Systemic inflammation-induced neuroinflammation is known to impair the autonomic center of cardiovascular regulation. The dynamic stability of blood pressure and heart rate (HR) is regulated by modulation of the reciprocal responses of sympathetic and parasympathetic tone by the baroreflex, which is controlled by the nucleus of the solitary tract (NTS). METHODS Systemic inflammation was induced by E. coli lipopolysaccharide (LPS, 1.2 mg/kg/day, 7 days) peritoneal infusion via an osmotic minipump in normotensive Sprague-Dawley rats. Systolic blood pressure (SBP) and HR were measured by femoral artery cannulation and recorded on a polygraph under anesthesia. The low-frequency (LF; 0.25-0.8 Hz) and high-frequency (HF; 0.8-2.4 Hz) components of SBP were adopted as the indices for sympathetic vasomotor tone and parasympathetic vasomotor tone, while the baroreflex effectiveness index (BEI) was adopted from the analysis of SBP and pulse interval (PI). The plasma levels of proinflammatory cytokines and mitochondrial DNA (mtDNA) oxidative damage were analyzed by ELISA. Protein expression was evaluated by Western blot. The distribution of oxidative mtDNA was probed by immunofluorescence. Pharmacological agents were delivered via infusion into the cisterna magna with an osmotic minipump. RESULTS The suppression of baroreflex sensitivity was concurrent with increased SBP and decreased HR. Neuroinflammatory factors, including TNF-α, CD11b, and Iba-1, were detected in the NTS of the LPS group. Moreover, indices of mtDNA damage, including 8-OHdG and γ-H2AX, were significantly increased in neuronal mitochondria. Pentoxifylline or minocycline intracisternal (IC) infusion effectively prevented mtDNA damage, suggesting that cytokine and microglial activation contributed to mtDNA damage. Synchronically, baroreflex sensitivity was effectively protected, and the elevated blood pressure was significantly relieved. In addition, the mtDNA repair mechanism was significantly enhanced by pentoxifylline or minocycline. CONCLUSION These results suggest that neuronal mtDNA damage in the NTS induced by neuroinflammation could be the core factor in deteriorating baroreflex desensitization and subsequent cardiovascular dysfunction. Therefore, the enhancement of base excision repair (BER) signaling in mitochondria could be a potential therapeutic strategy for cardiovascular reflex dysregulation.
Collapse
Affiliation(s)
- Mu-Hui Fu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan Republic of China
| | - I-Chun Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Chou-Hwei Lee
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Chih-Wei Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Yu-Chi Lee
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Yu Chih Kung
- Master of Science Program in Health Care, Department of Nursing, Meiho University, Neipu Township, Republic of China
- Department of Nursing, Meiho University, Neipu Township, Taiwan, Republic of China
| | - Chun-Ying Hung
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Kay L. H. Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
- Department of Senior Citizen Services, National Tainan Institute of Nursing, Tainan, 700 Taiwan Republic of China
| |
Collapse
|
111
|
Hypertension and Its Impact on Stroke Recovery: From a Vascular to a Parenchymal Overview. Neural Plast 2019; 2019:6843895. [PMID: 31737062 PMCID: PMC6815533 DOI: 10.1155/2019/6843895] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Hypertension is the first modifiable vascular risk factor accounting for 10.4 million deaths worldwide; it is strongly and independently associated with the risk of stroke and is related to worse prognosis. In addition, hypertension seems to be a key player in the implementation of vascular cognitive impairment. Long-term hypertension, complicated or not by the occurrence of ischemic stroke, is often reviewed on its vascular side, and parenchymal consequences are put aside. Here, we sought to review the impact of isolated hypertension or hypertension associated to stroke on brain atrophy, neuron connectivity and neurogenesis, and phenotype modification of microglia and astrocytes. Finally, we discuss the impact of antihypertensive therapies on cell responses to hypertension and functional recovery. This attractive topic remains a focus of continued investigation and stresses the relevance of including this vascular risk factor in preclinical investigations of stroke outcome.
Collapse
|
112
|
Li Y, Wei B, Liu X, Shen XZ, Shi P. Microglia, autonomic nervous system, immunity and hypertension: Is there a link? Pharmacol Res 2019; 155:104451. [PMID: 31557524 DOI: 10.1016/j.phrs.2019.104451] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/17/2019] [Accepted: 09/06/2019] [Indexed: 01/19/2023]
Abstract
Hypertension ranks the most common risk factor for cardiovascular diseases, and it affects almost one third of adult population globally. Emerging evidence indicates that immune activation is highly involved in the entire progress of hypertension and end organ damage. In addition to immunity, autonomic nervous system, particularly sympathetic nervous system, is one of the most conserved systems to maintain body homeostasis. Immune and sympathetic activities are found simultaneously increased in hypertension, suggesting a synergistic action of these two systems in the progression of this disease. Microglia, the primary immune cells in the central nervous system, have been suggested in the regulation of sympathetic outflow; depletion of microglia alters neuroinflammation and pressor responses in hypertensive models. In this review, we firstly updated the current understanding on microglial ontogeny and functions in both steady state and diseases. Then we reviewed on the interaction between autonomic nervous system and peripheral immunity in hypertension. Microglia bridge the central and peripheral inflammation via regulating the sympathetic nerve activity in hypertension. Future exploration of the molecular linkage of this pathway may provide novel therapeutic angel for hypertension and related cardiovascular diseases.
Collapse
Affiliation(s)
- You Li
- Department of Cardiology of the Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Medical Molecular Virology, Shanghai Medical College, Fudan University, China
| | - Bo Wei
- Department of Cardiology of the Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaoli Liu
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Xiao Z Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng Shi
- Department of Cardiology of the Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
113
|
Azzam O, Kiuchi MG, Ho JK, Matthews VB, Gavidia LML, Nolde JM, Carnagarin R, Schlaich MP. New Molecules for Treating Resistant Hypertension: a Clinical Perspective. Curr Hypertens Rep 2019; 21:80. [PMID: 31506798 DOI: 10.1007/s11906-019-0978-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW To review the findings of trials evaluating pharmacological treatment approaches for hypertension in general, and resistant hypertension (RH) in particular, and propose future research and clinical directions. RECENT FINDINGS RH is defined as blood pressure (BP) that remains above target levels despite adherence to at least three antihypertensive medications, including a diuretic. Thus far, clinical trials of pharmacological approaches in RH have focused on older molecules, with spironolactone being demonstrated as the most efficacious fourth-line agent. However, the use of spironolactone in clinical practice is hampered by its side effect profile and the risk of hyperkalaemia in important RH subgroups, such as patients with moderate-severe chronic kidney disease (CKD). Clinical trials of new molecules targeting both well-established and more recently elucidated pathophysiologic mechanisms of hypertension offer a multitude of potential treatment avenues that warrant further evaluation in the context of RH. These include selective mineralocorticoid receptor antagonists (MRAs), aldosterone synthase inhibitors (ASIs), activators of the counterregulatory renin-angiotensin-system (RAS), vaccines, neprilysin inhibitors alone and in combined formulations, natriuretic peptide receptor agonists A (NPRA-A) agonists, vasoactive intestinal peptide (VIP) agonists, centrally acting aminopeptidase A (APA|) inhibitors, antimicrobial suppression of central sympathetic outflow (minocycline), dopamine β-hydroxylase (DβH) inhibitors and Na+/H+ Exchanger 3 (NHE3) inhibitors. There is a paucity of data from trials evaluating newer molecules for the treatment of RH. Emergent novel molecules for non-resistant forms of hypertension heighten the prospects of identifying new, effective and well-tolerated pharmacological approaches to RH. There is a glaring need to undertake RH-focused trials evaluating their efficacy and clinical applicability.
Collapse
Affiliation(s)
- Omar Azzam
- Department of Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia.,Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Marcio G Kiuchi
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Jan K Ho
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Leslie Marisol Lugo Gavidia
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Janis M Nolde
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia. .,Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, Australia. .,Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
114
|
Yang T, Richards EM, Pepine CJ, Raizada MK. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol 2019; 14:442-456. [PMID: 29760448 DOI: 10.1038/s41581-018-0018-2] [Citation(s) in RCA: 469] [Impact Index Per Article: 78.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Crosstalk between the gut microbiota and the host has attracted considerable attention owing to its involvement in diverse diseases. Chronic kidney disease (CKD) is commonly associated with hypertension and is characterized by immune dysregulation, metabolic disorder and sympathetic activation, which are all linked to gut dysbiosis and altered host-microbiota crosstalk. In this Review, we discuss the complex interplay between the brain, the gut, the microbiota and the kidney in CKD and hypertension and explain our brain-gut-kidney axis hypothesis for the pathogenesis of these diseases. Consideration of the role of the brain-gut-kidney axis in the maintenance of normal homeostasis and of dysregulation of this axis in CKD and hypertension could lead to the identification of novel therapeutic targets. In addition, the discovery of unique microbial communities and their associated metabolites and the elucidation of brain-gut-kidney signalling are likely to fill fundamental knowledge gaps leading to innovative research, clinical trials and treatments for CKD and hypertension.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Elaine M Richards
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
115
|
Elsaafien K, Korim WS, Setiadi A, May CN, Yao ST. Chemoattraction and Recruitment of Activated Immune Cells, Central Autonomic Control, and Blood Pressure Regulation. Front Physiol 2019; 10:984. [PMID: 31427987 PMCID: PMC6688384 DOI: 10.3389/fphys.2019.00984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory mediators play a critical role in the regulation of sympathetic outflow to cardiovascular organs in hypertension. Emerging evidence highlights the involvement of immune cells in the regulation of blood pressure. However, it is still unclear how these immune cells are activated and recruited to key autonomic brain regions to regulate sympathetic outflow to cardiovascular organs. Chemokines such as C-C motif chemokine ligand 2 (CCL2), and pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β), are upregulated both peripherally and centrally in hypertension. More specifically, they are upregulated in key autonomic brain regions that control sympathetic activity and blood pressure such as the paraventricular nucleus of the hypothalamus and the rostral ventrolateral medulla. Furthermore, this upregulation of inflammatory mediators is associated with the infiltration of immune cells to these brain areas. Thus, expression of pro-inflammatory chemokines and cytokines is a potential mechanism promoting invasion of immune cells into key autonomic brain regions. In pathophysiological conditions, this can result in abnormal activation of brain circuits that control sympathetic nerve activity to cardiovascular organs and ultimately in increases in blood pressure. In this review, we discuss emerging evidence that helps explain how immune cells are chemoattracted to autonomic nuclei and contribute to changes in sympathetic outflow and blood pressure.
Collapse
Affiliation(s)
- Khalid Elsaafien
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Willian S. Korim
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Anthony Setiadi
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Clive N. May
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Song T. Yao
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
116
|
Myocardial Inflammation Predicts Remodeling and Neuroinflammation After Myocardial Infarction. J Am Coll Cardiol 2019; 71:263-275. [PMID: 29348018 DOI: 10.1016/j.jacc.2017.11.024] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/01/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The local inflammatory tissue response after acute myocardial infarction (MI) determines subsequent healing. Systemic interaction may induce neuroinflammation as a precursor to neurodegeneration. OBJECTIVES This study sought to assess the influence of MI on cardiac and brain inflammation using noninvasive positron emission tomography (PET) of the heart-brain axis. METHODS After coronary artery ligation or sham surgery, mice (n = 49) underwent serial whole-body PET imaging of the mitochondrial translocator protein (TSPO) as a marker of activated macrophages and microglia. Patients after acute MI (n = 3) were also compared to healthy controls (n = 9). RESULTS Infarct mice exhibited elevated myocardial TSPO signal at 1 week versus sham (percent injected dose per gram: 8.0 ± 1.6 vs. 4.8 ± 0.9; p < 0.001), localized to activated CD68+ inflammatory cells in the infarct. Early TSPO signal predicted subsequent left ventricular remodeling at 8 weeks (rpartial = -0.687; p = 0.001). In parallel, brain TSPO signal was elevated at 1 week (1.7 ± 0.2 vs. 1.4 ± 0.2 for sham; p = 0.017), localized to activated microglia. After interval decline at 4 weeks, progressive heart failure precipitated a second wave of neuroinflammation (1.8 ± 0.2; p = 0.005). TSPO was concurrently up-regulated in remote cardiomyocytes at 8 weeks (8.8 ± 1.7, p < 0.001) without inflammatory cell infiltration, suggesting mitochondrial impairment. Angiotensin-converting enzyme inhibitor treatment lowered acute inflammation in the heart (p = 0.003) and brain (p = 0.06) and improved late cardiac function (p = 0.05). Patients also demonstrated elevation of cardiac TSPO signal in the infarct territory, paralleled by neuroinflammation versus controls. CONCLUSIONS The brain is susceptible to acute MI and chronic heart failure. Immune activation may interconnect heart and brain dysfunction, a finding that provides a foundation for strategies to improve heart and brain outcomes.
Collapse
|
117
|
Abstract
Purpose of Review Hypertension is related to impaired metabolic homeostasis and can be regarded as a metabolic disorder. This review presents possible mechanisms by which metabolic disorders increase blood pressure (BP) and discusses the importance of the gut as a novel modulator of BP. Recent Findings Obesity and high salt intake are major risk factors for hypertension. There is a hypothesis of “salt-induced obesity”; i.e., high salt intake may tie to obesity. Heightened sympathetic nervous system (SNS) activity, especially in the kidney and brain, increases BP in obese patients. Adipokines, including adiponectin and leptin, and renin-angiotensin-aldosterone system (RAAS) contribute to hypertension. Adiponectin induced by a high-salt diet may decrease sodium/glucose cotransporter (SGLT) 2 expression in the kidney, which results in reducing BP. High salt can change secretions of adipokines and RAAS-related components. Evidence has been accumulating linking the gastrointestinal tract to BP. Glucagon-like peptide-1 (GLP-1) and ghrelin decrease BP in both rodents and humans. The sweet taste receptor in enteroendocrine cells increases SGLT1 expression and stimulates sodium/glucose absorption. Roux-en-Y gastric bypass improves glycemic and BP control due to reducing the activity of SGLT1. Na/H exchanger isoform 3 (NHE3) increases BP by stimulating the intestinal absorption of sodium. Gastrin functions as an intestinal sodium taste sensor and inhibits NHE3 activity. Intestinal mineralocorticoid receptors also regulate sodium absorption and BP due to changing ENaC activity. Gastric sensing of sodium induces natriuresis, and gastric distension increases BP. Changes in the composition and function of gut microbiota contribute to hypertension. A high-salt/fat diet may disrupt the gut barrier, which results in systemic inflammation, insulin resistance, and increased BP. Gut microbiota regulates BP by secreting vasoactive hormones and short-chain fatty acids. BP-lowering effects of probiotics and antibiotics have been reported. Bariatric surgery improves metabolic disorders and hypertension due to increasing GLP-1 secretion, decreasing leptin secretion and SNS activity, and changing gut microbiome composition. Strategies targeting the gastrointestinal system may be therapeutic options for improving metabolic abnormalities and reducing BP in humans. Summary SNS, brain, adipocytes, RAAS, the kidney, the gastrointestinal tract, and microbiota play important roles in regulating BP. Most notably, the gut could be a novel target for treatment of hypertension as a metabolic disorder.
Collapse
|
118
|
Wang HW, Ahmad M, Jadayel R, Najjar F, Lagace D, Leenen FHH. Inhibition of inflammation by minocycline improves heart failure and depression-like behaviour in rats after myocardial infarction. PLoS One 2019; 14:e0217437. [PMID: 31233508 PMCID: PMC6590948 DOI: 10.1371/journal.pone.0217437] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/10/2019] [Indexed: 11/29/2022] Open
Abstract
RATIONALE Patients with heart failure have an increased incidence of depression. Central and peripheral inflammation play a major role in the pathophysiology of both heart failure and depression. AIM Minocycline is an antibiotic that inhibits microglia activation and release of pro-inflammatory cytokines. We assessed effects of minocycline on extent of heart failure and depression at 2 and 8 weeks post myocardial infarction. METHODS/RESULTS Male Wistar rats were randomly divided into 3 groups: (i) sham + vehicle; (ii) MI + vehicle; and (iii) MI + minocycline with n/group of 8, 9 and 9 at 2 weeks, and 10, 16, 8 at weeks, respectively. Oral minocycline (50 mg/kg/day) or vehicle started 2 days before surgery. Depression-like behaviour was assessed with sucrose preference and forced swim tests, and cardiac function with echo and hemodynamics. After myocardial infarction, microglia activation and plasma/brain pro-inflammatory cytokines increased, which were mostly prevented by minocycline. At 8 weeks, cardiac dysfunction was attenuated by minocycline: infarct size (MI + Vehicle 29±1, MI + Min 23±1%), ejection fraction (Sham 80±1, MI + Vehicle 48±2, MI + Min 58±2%) and end diastolic pressure (Sham 3.2±0.3, MI + Vehicle 18.2±1.1, MI + Min 8.5±0.9 mm Hg). Depression-like behaviour was significantly improved by minocycline in sucrose preference test (% Sucrose Intake: Sham 96±1, MI + Vehicle 78±2, MI + Min 87±2) and forced swim test (% Immobile: Sham 40±4, MI + Vehicle 61±3, MI + Min 37±6). CONCLUSION Rats post myocardial infarction develop systemic inflammation, heart failure and depression-like behaviour that are all attenuated by minocycline. Targeting (neuro) inflammation may represent new therapeutic strategy for patients with heart failure and depression.
Collapse
Affiliation(s)
- Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Rami Jadayel
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Fatimah Najjar
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Diane Lagace
- Neuroscience Research Program, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Frans H. H. Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| |
Collapse
|
119
|
Elkhatib SK, Case AJ. Autonomic regulation of T-lymphocytes: Implications in cardiovascular disease. Pharmacol Res 2019; 146:104293. [PMID: 31176794 DOI: 10.1016/j.phrs.2019.104293] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/22/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022]
Abstract
The nervous and immune systems both serve as essential assessors and regulators of physiological function. Recently, there has been a great interest in how the nervous and immune systems interact to modulate both physiological and pathological states. In particular, the autonomic nervous system has a direct line of communication with immune cells anatomically, and moreover, immune cells possess receptors for autonomic neurotransmitters. This circumstantial evidence is suggestive of a functional interplay between the two systems, and extensive research over the past few decades has demonstrated neurotransmitters such as the catecholamines (i.e. dopamine, norepinephrine, and epinephrine) and acetylcholine have potent immunomodulating properties. Furthermore, immune cells, particularly T-lymphocytes, have now been found to express the cellular machinery for both the synthesis and degradation of neurotransmitters, which suggests the ability for both autocrine and paracrine signaling from these cells independent of the nervous system. The details underlying the functional interplay of this complex network of neuroimmune communication are still unclear, but this crosstalk is suggestive of significant implications on the pathogenesis of a number of autonomic-dysregulated and inflammation-mediated diseases. In particular, it is widely accepted that numerous forms of cardiovascular diseases possess imbalanced autonomic tone as well as altered T-lymphocyte function, but a paucity of literature exists discussing the direct role of neurotransmitters in shaping the inflammatory microenvironment during the progression or therapeutic management of these diseases. This review seeks to provide a fundamental framework for this autonomic neuroimmune interaction within T-lymphocytes, as well as the implications this may have in cardiovascular diseases.
Collapse
Affiliation(s)
- Safwan K Elkhatib
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Adam J Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
120
|
Wang S, Wu L, Zhai Y, Li X, Li B, Zhao D, Jiang H. Noninvasive light emitting diode therapy: A novel approach for postinfarction ventricular arrhythmias and neuroimmune modulation. J Cardiovasc Electrophysiol 2019; 30:1138-1147. [PMID: 31104349 DOI: 10.1111/jce.13974] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sympathetic neural activation plays a key role in the incidence and maintenance of acute myocardial infarction (AMI) induced ventricular arrhythmia (VA). Furthermore, previous studies showed that AMI might induce microglia and sympathetic activation and that microglial activation might contribute to sympathetic activation. Recently, studies showed that light emitting diode (LED) therapy might attenuate microglial activation. Therefore, we hypothesized that LED therapy might reduce AMI-induced VA by attenuating microglia and sympathetic activation. METHODS Thirty anesthetized rats were randomly divided into three groups: the Control group (n = 6), AMI group (n = 12), and AMI + LED group (n = 12). Electrocardiogram (ECG) and left stellate ganglion (LSG) neural activity were continuously recorded. The incidence of VAs was recorded during the first hour after AMI. Furthermore, we sampled the brain and myocardium tissue of the different groups to examine the microglial activation and expression of nerve growth factor (NGF), interleukin-18 (IL-18), and IL-1β, respectively. RESULTS Compared to the AMI group, LED therapy significantly reduced the incidence of AMI-induced VAs (ventricular premature beats [VPB] number: 85.08 ± 13.91 vs 27.5 ± 9.168, P < .01; nonsustained ventricular tachycardia (nSVT) duration: 34.39 ± 8.562 vs 9.005 ± 3.442, P < .05; nSVT number: 18.92 ± 4.52 vs 7.583 ± 3.019, P < .05; incidence rate of SVT/VF: 58.33% vs. 8.33%, P < .05) and reduced the LSG neural activity (P < .01) in the AMI + LED group. Furthermore, LED significantly attenuated microglial activation and reduced IL-18, IL-1β, and NGF expression in the peri-infarct myocardium. CONCLUSION LED therapy may protect against AMI-induced VAs by suppressing sympathetic neural activity and the inflammatory response.
Collapse
Affiliation(s)
- Songyun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Lin Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yi Zhai
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Xuemeng Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Binxun Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Dongdong Zhao
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
121
|
Carnagarin R, Matthews V, Zaldivia MTK, Peter K, Schlaich MP. The bidirectional interaction between the sympathetic nervous system and immune mechanisms in the pathogenesis of hypertension. Br J Pharmacol 2019; 176:1839-1852. [PMID: 30129037 PMCID: PMC6534787 DOI: 10.1111/bph.14481] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022] Open
Abstract
Over the last few years, evidence has accumulated to suggest that hypertension is, at least in part, an immune-mediated inflammatory disorder. Many links between immunity and hypertension have been established and provide a complex framework of mechanistic interactions contributing to the rise in BP. These include immune-mediated inflammatory processes affecting regulatory brain nuclei and interactions with other mediators of cardiovascular regulation such as the sympathetic nervous system. Sympathoexcitation differentially regulates T-cells based upon activation status of the immune cell as well as the resident organ. Exogenous and endogenous triggers activate signalling pathways in innate and adaptive immune cells resulting in pro-inflammatory cytokine production and activation of T-lymphocytes in the cardiovascular and renal regions, now considered major factors in the development of essential hypertension. The inflammatory cascade is sustained and exacerbated by the immune flow via the brain-bone marrow-spleen-gastrointestinal axis and thereby further aggravating immune-mediated pathways resulting in a vicious cycle of established hypertension and target organ damage. This review summarizes the evidence and recent advances in linking immune-mediated inflammation, sympathetic activation and their bidirectional interactions with the development of hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
| | - Vance Matthews
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
| | - Maria T K Zaldivia
- Atherothrombosis and Vascular BiologyBaker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of MedicineMonash University, Royal Perth HospitalPerthWAAustralia
| | - Karlheinz Peter
- Atherothrombosis and Vascular BiologyBaker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of MedicineMonash University, Royal Perth HospitalPerthWAAustralia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
- Department of CardiologyRoyal Perth HospitalPerthWAAustralia
- Department of NephrologyRoyal Perth HospitalPerthWAAustralia
| |
Collapse
|
122
|
Chan JYH, Chan SHH. Differential impacts of brain stem oxidative stress and nitrosative stress on sympathetic vasomotor tone. Pharmacol Ther 2019; 201:120-136. [PMID: 31153955 DOI: 10.1016/j.pharmthera.2019.05.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Abstract
Based on work-done in the rostral ventrolateral medulla (RVLM), this review presents four lessons learnt from studying the differential impacts of oxidative stress and nitrosative stress on sympathetic vasomotor tone and their clinical and therapeutic implications. The first lesson is that an increase in sympathetic vasomotor tone because of augmented oxidative stress in the RVLM is responsible for the generation of neurogenic hypertension. On the other hand, a shift from oxidative stress to nitrosative stress in the RVLM underpins the succession of increase to decrease in sympathetic vasomotor tone during the progression towards brain stem death. The second lesson is that, by having different cellular sources, regulatory mechanisms on synthesis and degradation, kinetics of chemical reactions, and downstream signaling pathways, reactive oxygen species and reactive nitrogen species should not be regarded as a singular moiety. The third lesson is that well-defined differential roles of oxidative stress and nitrosative stress with distinct regulatory mechanisms in the RVLM during neurogenic hypertension and brain stem death clearly denote that they are not interchangeable phenomena with unified cellular actions. Special attention must be paid to their beneficial or detrimental roles under a specific disease or a particular time-window of that disease. The fourth lesson is that, to be successful, future antioxidant therapies against neurogenic hypertension must take into consideration the much more complicated picture than that presented in this review on the generation, maintenance, regulation or modulation of the sympathetic vasomotor tone. The identification that the progression towards brain stem death entails a shift from oxidative stress to nitrosative stress in the RVLM may open a new vista for therapeutic intervention to slow down this transition.
Collapse
Affiliation(s)
- Julie Y H Chan
- Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan, Republic of China
| | - Samuel H H Chan
- Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan, Republic of China.
| |
Collapse
|
123
|
Wu Q, Mi Y, Cheng W, Xia C, Zhu D, Du D. Infiltrating T helper 17 cells in the paraventricular nucleus are pathogenic for stress-induced hypertension. Biochem Biophys Res Commun 2019; 515:169-175. [PMID: 31133377 DOI: 10.1016/j.bbrc.2019.05.121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/19/2019] [Indexed: 01/04/2023]
Abstract
Central neuroinflammation produced by both innate and adaptive immunities plays a major role in the development of stress-induced hypertension (SIH), but successful T cell immunoregulation for SIH requires that the T cells can access brain tissues. So far, both the effects of T helper 17 (Th17) cells on SIH and the pathway for T cells entry into the brain were unknown. Here we show that the blood pressure (BP), heart rate (HR) and the norepinephrine(NE) of the SIH rats were considerably higher, the numbers of Th17 cells and IL-17 were higher, relative to control. Anti-IL-17 attenuated the elevation of BP and HR of the SIH rats when microinjected into the paraventricular nucleus (PVN).Alb-FITC, after infusion into the carotid artery, were found in the brain parenchyma of the PVN in the SIH rats. We concluded that Th17 cells infiltrated the parenchyma of the paraventricular nucleus (PVN) via a compromised blood brain barrier (BBB) in response to stress and Th17 cells and IL-17 play an important role in the pathophysiology of SIH.
Collapse
Affiliation(s)
- Qin Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yang Mi
- School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Wenjing Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Danian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| | - Dongshu Du
- School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
124
|
Liu J, Wu J. Tumour necrosis factor α sets area postrema on fire in renovascular hypertension. Cardiovasc Res 2019; 115:995-997. [PMID: 30657874 DOI: 10.1093/cvr/cvz011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jing Liu
- Department of Physiology, The Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, USA
| | - Jing Wu
- Department of Physiology, The Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, USA
| |
Collapse
|
125
|
Oliveira AC, Sharma RK, Aquino V, Lobaton G, Bryant AJ, Harrison JK, Richards EM, Raizada MK. Involvement of Microglial Cells in Hypoxia-induced Pulmonary Hypertension. Am J Respir Cell Mol Biol 2019; 59:271-273. [PMID: 30067089 DOI: 10.1165/rcmb.2018-0042le] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Aline C Oliveira
- 1 University of Florida College of Medicine Gainesville, Florida
| | | | - Victor Aquino
- 1 University of Florida College of Medicine Gainesville, Florida
| | - Gilberto Lobaton
- 1 University of Florida College of Medicine Gainesville, Florida
| | - Andrew J Bryant
- 1 University of Florida College of Medicine Gainesville, Florida
| | | | | | - Mohan K Raizada
- 1 University of Florida College of Medicine Gainesville, Florida
| |
Collapse
|
126
|
Carvalho-Galvão A, Guimarães DD, De Brito Alves JL, Braga VA. Central Inhibition of Tumor Necrosis Factor Alpha Reduces Hypertension by Attenuating Oxidative Stress in the Rostral Ventrolateral Medulla in Renovascular Hypertensive Rats. Front Physiol 2019; 10:491. [PMID: 31114507 PMCID: PMC6502978 DOI: 10.3389/fphys.2019.00491] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation in the central nervous system is being considered a key player linked to neurogenic hypertension. Using combined in vivo and in vitro approaches, we investigated the effects of central inhibition of TNF-α on blood pressure, sympathetic tone, baroreflex sensitivity, and oxidative stress in the rostral ventrolateral medulla (RVLM) of rats with 2-kidney-1-clip (2K1C) renovascular hypertension. Continuous infusion of pentoxifylline, a TNF-α inhibitor, into the lateral ventricle of the brain for 14 consecutive days reduced blood pressure and improved baroreflex sensitivity in renovascular hypertensive rats. Furthermore, central TNF-α inhibition reduced sympathetic modulation and blunted the increased superoxide accumulation in the RVLM of 2K1C rats. Our findings suggest that TNF-α play an important role in the maintenance of sympathetic vasomotor tone and increased oxidative stress in the RVLM during renovascular hypertension.
Collapse
Affiliation(s)
| | - Drielle D Guimarães
- Biotechnology Center, Federal University of Paraíba, João Pessoa, Brazil.,Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - José L De Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil
| | - Valdir A Braga
- Biotechnology Center, Federal University of Paraíba, João Pessoa, Brazil.,Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
127
|
Mowry FE, Biancardi VC. Neuroinflammation in hypertension: the renin-angiotensin system versus pro-resolution pathways. Pharmacol Res 2019; 144:279-291. [PMID: 31039397 DOI: 10.1016/j.phrs.2019.04.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/31/2022]
Abstract
Overstimulation of the pro-inflammatory pathways within brain areas responsible for sympathetic outflow is well evidenced as a primary contributing factor to the establishment and maintenance of neurogenic hypertension. However, the precise mechanisms and stimuli responsible for promoting a pro-inflammatory state are not fully elucidated. Recent work has unveiled novel compounds derived from omega-3 polyunsaturated fatty acids (ω-3 PUFAs), termed specialized pro-resolving mediators (SPMs), which actively regulate the resolution of inflammation. Failure or dysregulation of the resolution process has been linked to a variety of chronic inflammatory and neurodegenerative diseases. Given the pathologic role of neuroinflammation in the hypertensive state, SPMs and their associated pathways may provide a link between hypertension and the long-standing association of dietary ω-3 PUFAs with cardioprotection. Herein, we review recent progress in understanding the RAS-driven pathophysiology of neurogenic hypertension, particularly in regards to the chronic low-grade neuroinflammatory response. In addition, we examine the potential for an impaired resolution of inflammation process in the context of hypertension.
Collapse
Affiliation(s)
- Francesca Elisabeth Mowry
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Alabama, USA
| | - Vinicia Campana Biancardi
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Alabama, USA; Center for Neurosciences Research Initiative, Auburn University, Alabama, USA.
| |
Collapse
|
128
|
AlMarabeh S, Abdulla MH, O'Halloran KD. Is Aberrant Reno-Renal Reflex Control of Blood Pressure a Contributor to Chronic Intermittent Hypoxia-Induced Hypertension? Front Physiol 2019; 10:465. [PMID: 31105584 PMCID: PMC6491928 DOI: 10.3389/fphys.2019.00465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/04/2019] [Indexed: 12/16/2022] Open
Abstract
Renal sensory nerves are important in the regulation of body fluid and electrolyte homeostasis, and blood pressure. Activation of renal mechanoreceptor afferents triggers a negative feedback reno-renal reflex that leads to the inhibition of sympathetic nervous outflow. Conversely, activation of renal chemoreceptor afferents elicits reflex sympathoexcitation. Dysregulation of reno-renal reflexes by suppression of the inhibitory reflex and/or activation of the excitatory reflex impairs blood pressure control, predisposing to hypertension. Obstructive sleep apnoea syndrome (OSAS) is causally related to hypertension. Renal denervation in patients with OSAS or in experimental models of chronic intermittent hypoxia (CIH), a cardinal feature of OSAS due to recurrent apnoeas (pauses in breathing), results in a decrease in circulating norepinephrine levels and attenuation of hypertension. The mechanism of the beneficial effect of renal denervation on blood pressure control in models of CIH and OSAS is not fully understood, since renal denervation interrupts renal afferent signaling to the brain and sympathetic efferent signals to the kidneys. Herein, we consider the currently proposed mechanisms involved in the development of hypertension in CIH disease models with a focus on oxidative and inflammatory mediators in the kidneys and their potential influence on renal afferent control of blood pressure, with wider consideration of the evidence available from a variety of hypertension models. We draw focus to the potential contribution of aberrant renal afferent signaling in the development, maintenance and progression of high blood pressure, which may have relevance to CIH-induced hypertension.
Collapse
Affiliation(s)
- Sara AlMarabeh
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Mohammed H Abdulla
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
129
|
Guyenet PG, Stornetta RL, Holloway BB, Souza GMPR, Abbott SBG. Rostral Ventrolateral Medulla and Hypertension. Hypertension 2019; 72:559-566. [PMID: 30354763 DOI: 10.1161/hypertensionaha.118.10921] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Patrice G Guyenet
- From the Department of Pharmacology, University of Virginia, Charlottesville
| | - Ruth L Stornetta
- From the Department of Pharmacology, University of Virginia, Charlottesville
| | - Benjamin B Holloway
- From the Department of Pharmacology, University of Virginia, Charlottesville
| | - George M P R Souza
- From the Department of Pharmacology, University of Virginia, Charlottesville
| | - Stephen B G Abbott
- From the Department of Pharmacology, University of Virginia, Charlottesville
| |
Collapse
|
130
|
Meng L, Bai X, Zheng Y, Chen D, Zheng Y. Altered expression of norepinephrine transporter participate in hypertension and depression through regulated TNF-α and IL-6. Clin Exp Hypertens 2019; 42:181-189. [PMID: 30957546 DOI: 10.1080/10641963.2019.1601205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aim: We explored the role of histone modification in the association of depression-hypertension by comparing norepinephrine transporter (NET) gene levels in different depression-hypertensive patients. Then, we analyzed the expression of NET correlation with inflammatory cytokines to provide a new direction for detecting the association mechanism between depression and hypertension.Methods: NE expression levels in serum of diverse groups were detected by enzyme-linked immunosorbent assay. Then histone acetyltransferase (HAT), histone deacetylase (HDAC), H3K27ac, NET, TNF-α, and interleukin-6 (IL-6) were detected by western blot in nine female subjects in different depression and hypertension groups, and Chromatin immunoprecipitation-polymerase chain reaction (Chip-PCR) were used to confirm the degree of acetylation affecting on the transcription level of NET gene. Meanwhile, correlation between NET with TNF/IL-6 was analyzed by SPSS19.0 software program. Finally, Quantitative real-time polymerase chain reaction (qPCR) and western blot were used to detect TNF-α and IL-6 expression levels after NET overexpression or interference treatment in human umbilical vein endothelial cells and Neuro-2a cells.Results: The expression of HAT and H3K27ac had lower levels in D-H and nonD-H group than nonD-nonH group. The results showed that higher acetylation could promote expression of NET genes. Meanwhile, the expression of NET had a significant negative correlation with IL-6 (R = -0.933, p < 0.01) and tumor necrosis factor (TNF) (R = -0.817, p < 0.01) in subjects. In addition, the results confirmed that TNF-α and IL-6 mRNA and protein partial expressions could be inhibited by NET in both HUVECs and Neuronal cells (p < 0.01).Conclusion: In conclusion, differential expression of NET gene might function as an important factor in interaction between depression and hypertension by partially targeting TNF-α and IL-6.
Collapse
Affiliation(s)
- Lin Meng
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Xiaoxue Bai
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Yan Zheng
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Dongmei Chen
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Yang Zheng
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| |
Collapse
|
131
|
Affiliation(s)
- Rhian M Touyz
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Livia L Camargo
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| |
Collapse
|
132
|
Żera T, Nowiński A, Segiet A, Smykiewicz P. Microglia and brain angiotensin type 1 receptors are involved in desensitising baroreflex by intracerebroventricular hypertonic saline in male Sprague-Dawley rats. Auton Neurosci 2019; 217:49-57. [PMID: 30704975 DOI: 10.1016/j.autneu.2019.01.002] [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: 09/06/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 12/09/2022]
Abstract
High salt diet alters cardiovascular control by increasing concentration of sodium ions (Na+) in cerebrospinal fluid (CSF) and is a risk factor for hypertension. Hypernatremic conditions activate microglia and upregulate renin-angiotensin system in the brain. Thus, we checked if chronic elevation of CSF Na+ affects neural control of circulatory system via microglia and brain angiotensin type 1 receptors (AT1Rs). Normotensive adult male Sprague-Dawley rats received two-week intracerebroventricular (ICV) infusion of either isoosmotic saline (0.9% NaCl); hyperosmotic saline (5% NaCl); 5% NaCl with minocycline - inhibitor of microglia; 5% NaCl with losartan - AT1R blocker. Fluid intake, urine output, and urinary Na+ excretion were measured before and during ICV infusions. At the end of ICV infusions, blood pressure and heart rate were recorded in awake rats at rest, in response to acute air jet stressor, during pharmacological evaluation of baroreflex, and after autonomic ganglia blockade. CSF and blood were collected for evaluation of Na+ concentration. Baroreflex was blunted in rats ICV infused with 5% NaCl. ICV treatment with losartan or minocycline prevented decrease in baroreflex sensitivity. Hemodynamic parameters at rest, in response to acute stressor and autonomic ganglia blockade were similar in all groups. Neither treatment affected water intake, urine output and urinary Na+ excretion. ICV infusion of 5% NaCl resulted in higher concentration of Na+ in CSF than in control group (0.9% NaCl) and in plasma. Our results indicate that chronic ICV infusion of hyperosmotic saline blunts baroreflex in normotensive rats and this desensitization is mediated by microglia and AT1Rs.
Collapse
Affiliation(s)
- Tymoteusz Żera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, the Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland.
| | - Artur Nowiński
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, the Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Agnieszka Segiet
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, the Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Paweł Smykiewicz
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, the Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| |
Collapse
|
133
|
Nakayama Y, Fujiu K. Maladaptive Alterations of Autonomic Nerve System in Cardiovascular Disorders. Int Heart J 2019; 60:4-6. [DOI: 10.1536/ihj.18-677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Yukiteru Nakayama
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Katsuhito Fujiu
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
- Department of Advanced Cardiology, The University of Tokyo
| |
Collapse
|
134
|
Małkiewicz MA, Szarmach A, Sabisz A, Cubała WJ, Szurowska E, Winklewski PJ. Blood-brain barrier permeability and physical exercise. J Neuroinflammation 2019; 16:15. [PMID: 30678702 PMCID: PMC6345022 DOI: 10.1186/s12974-019-1403-x] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
In this narrative review, a theoretical framework on the crosstalk between physical exercise and blood-brain barrier (BBB) permeability is presented. We discuss the influence of physical activity on the factors affecting BBB permeability such as systemic inflammation, the brain renin-angiotensin and noradrenergic systems, central autonomic function and the kynurenine pathway. The positive role of exercise in multiple sclerosis and Alzheimer’s disease is described. Finally, the potential role of conditioning as well as the effect of exercise on BBB tight junctions is outlined. There is a body of evidence that regular physical exercise diminishes BBB permeability as it reinforces antioxidative capacity, reduces oxidative stress and has anti-inflammatory effects. It improves endothelial function and might increase the density of brain capillaries. Thus, physical training can be emphasised as a component of prevention programs developed for patients to minimise the risk of the onset of neuroinflammatory diseases as well as an augmentation of existing treatment. Unfortunately, despite a sound theoretical background, it remains unclear as to whether exercise training is effective in modulating BBB permeability in several specific diseases. Further research is needed as the impact of exercise is yet to be fully elucidated.
Collapse
Affiliation(s)
- Marta A Małkiewicz
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland.,Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Arkadiusz Szarmach
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Sabisz
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Wiesław J Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Edyta Szurowska
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Paweł J Winklewski
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland. .,2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland. .,Department of Clinical Anatomy and Physiology, Faculty of Health Sciences, Pomeranian University of Slupsk, Slupsk, Poland.
| |
Collapse
|
135
|
|
136
|
Importance of the commissural nucleus of the solitary tract in renovascular hypertension. Hypertens Res 2019; 42:587-597. [PMID: 30622315 DOI: 10.1038/s41440-018-0190-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 12/12/2022]
Abstract
The rodent renovascular hypertension model has been used to investigate the mechanisms promoting hypertension. The importance of the carotid body for renovascular hypertension has been demonstrated. As the commissural NTS (cNTS) is the first synaptic site in the central nervous system that receives information from carotid body chemoreceptors, we evaluated the contribution of cNTS to renovascular hypertension in the present study. Normotensive male Holtzman rats were implanted with a silver clip around the left renal artery to induce two-kidney, one-clip (2K1C) hypertension. Six weeks later, isoguvacine (a GABAA agonist) or losartan (an AT1 antagonist) was injected into the cNTS, and the effects were compared with carotid body removal. Immunohistochemistry for Iba-1 and GFAP to label microglia and astrocytes, respectively, and RT-PCR for components of the renin-angiotensin system and cytokines in the NTS were also performed 6 weeks after renal surgery. The inhibition of cNTS with isoguvacine or the blockade of AT1 receptors with losartan in the cNTS decreased the blood pressure and heart rate of 2K1C rats even more than carotid body removal did. The mRNA expression of NOX2, TNF-α and IL-6, microglia, and astrocytes also increased in the cNTS of 2K1C rats compared to that of normotensive rats. These results indicate that tonically active neurons within the cNTS are essential for the maintenance of hypertension in 2K1C rats. In addition to signals from the carotid body, the present results suggest that angiotensin II directly activates the cNTS and may also induce microgliosis and astrogliosis within the NTS, which, in turn, cause oxidative stress and neuroinflammation.
Collapse
|
137
|
Segiet A, Smykiewicz P, Kwiatkowski P, Żera T. Tumour necrosis factor and interleukin 10 in blood pressure regulation in spontaneously hypertensive and normotensive rats. Cytokine 2019; 113:185-194. [DOI: 10.1016/j.cyto.2018.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/30/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023]
|
138
|
Wen Y, Crowley SD. Renal Effects of Cytokines in Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:443-454. [PMID: 31399978 DOI: 10.1007/978-981-13-8871-2_21] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Preclinical studies point to a key role for immune cells in hypertension via augmenting renal injury and/or hypertensive responses. Blood pressure elevation in rheumatologic patients is attenuated by anti-inflammatory therapies. Both the innate and adaptive immune systems contribute to the pathogenesis of hypertension by modulating renal sodium balance, blood flow, and functions of the vasculature and epithelial cells in the kidney. Monocytes/macrophages and T lymphocytes are pivotal mediators of hypertensive responses, while dendritic cells and B lymphocytes can regulate blood pressure indirectly by promoting T lymphocytes activation. Pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF), interleukin-1 (IL-1), interleukin-17 (IL-17), and interferon-γ (IFN), amplify blood pressure elevation and/or renal injury. By contrast, interleukin-10 (IL-10) protects against renal and vascular function when produced by T helper 2 cells (Th2) and regulatory T cells (Treg). Thus, understanding the renal effects of cytokines in hypertension will provide targets for precise immunotherapies to inhibit targeted organ damage while preserving necessary immunity.
Collapse
Affiliation(s)
- Yi Wen
- Division of Nephrology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China.,Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA.
| |
Collapse
|
139
|
Cohen EM, Mohammed S, Kavurma M, Nedoboy PE, Cartland S, Farnham MM, Pilowsky PM. Microglia in the RVLM of SHR have reduced P2Y12R and CX3CR1 expression, shorter processes, and lower cell density. Auton Neurosci 2019; 216:9-16. [DOI: 10.1016/j.autneu.2018.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 01/06/2023]
|
140
|
Basting T, Xu J, Mukerjee S, Epling J, Fuchs R, Sriramula S, Lazartigues E. Glutamatergic neurons of the paraventricular nucleus are critical contributors to the development of neurogenic hypertension. J Physiol 2018; 596:6235-6248. [PMID: 30151830 PMCID: PMC6292814 DOI: 10.1113/jp276229] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/17/2018] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Recurrent periods of over-excitation in the paraventricular nucleus (PVN) of the hypothalamus could contribute to chronic over-activation of this nucleus and thus enhanced sympathetic drive. Stimulation of the PVN glutamatergic population utilizing channelrhodopsin-2 leads to an immediate frequency-dependent increase in baseline blood pressure. Partial lesions of glutamatergic neurons of the PVN (39.3%) result in an attenuated rise in blood pressure following Deoxycorticosterone acetate (DOCA)-salt treatment and reduced index of sympathetic activity. These data suggest that stimulation of PVN glutamatergic neurons is sufficient to cause autonomic dysfunction and drive the increase in blood pressure during hypertension. ABSTRACT Neuro-cardiovascular dysregulation leads to increased sympathetic activity and neurogenic hypertension. The paraventricular nucleus (PVN) of the hypothalamus is a key hub for blood pressure (BP) control, producing or relaying the increased sympathetic tone in hypertension. We hypothesize that increased central sympathetic drive is caused by chronic over-excitation of glutamatergic PVN neurons. We tested how stimulation or lesioning of excitatory PVN neurons in conscious mice affects BP, baroreflex and sympathetic activity. Glutamatergic PVN neurons were unilaterally transduced with channelrhodopsin-2 using an adeno-associated virus (CamKII-ChR2-eYFP-AAV2) in wildtype mice (n = 7) to assess the impact of acute stimulation of excitatory PVN neurons selectively on resting BP in conscious mice. Stimulation of the PVN glutamatergic population resulted in an immediate frequency-dependent (2, 10 and 20 Hz) increase in BP from baseline by ∼9 mmHg at 20 Hz stimulation (P < 0.001). Additionally, in vGlut2-cre mice glutamatergic neurons of the PVN were bilaterally lesioned utilizing a cre-dependent caspase (AAV2-flex-taCASP3-TEVp). Resting BP and urinary noradrenaline (norepinephrine) levels were then recorded in conscious mice before and after DOCA-salt hypertension. Partial lesions of glutamatergic neurons of the PVN (39.3%, P < 0.05) resulted in an attenuated rise in BP following DOCA-salt treatment (P < 0.05 at 7 day time point, n = 8). Noradrenaline levels as an index of sympathetic activity between the lesion and wildtype groups showed a significant reduction after DOCA-salt treatment in the lesioned animals (P < 0.05). These experiments suggest that stimulation of PVN glutamatergic neurons is sufficient to cause autonomic dysfunction and drive the increase in BP.
Collapse
Affiliation(s)
- Tyler Basting
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
| | - Jiaxi Xu
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
| | - Snigdha Mukerjee
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
| | - Joel Epling
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
| | - Robert Fuchs
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
| | - Srinivas Sriramula
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
- Department of Pharmacology and Toxicology, Brody School of MedicineEast Carolina UniversityGreenvilleNC27834USA
| | - Eric Lazartigues
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA70112USA
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA70112USA
- Neuroscience Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA70112USA
- Southeast Louisiana Veterans Health Care SystemNew OrleansLAUSA
| |
Collapse
|
141
|
Abstract
The causes of essential hypertension remain an enigma. Interactions between genetic and external factors are generally recognized to act as aetiological mechanisms that trigger the pathogenesis of high blood pressure. However, the questions of which genes and factors are involved, and when and where such interactions occur, remain unresolved. Emerging evidence indicates that the hypertensive response to pressor stimuli, like many other physiological and behavioural adaptations, can become sensitized to particular stimuli. Studies in animal models show that, similarly to other response systems controlled by the brain, hypertensive response sensitization (HTRS) is mediated by neuroplasticity. The brain circuitry involved in HTRS controls the sympathetic nervous system. This Review outlines evidence supporting the phenomenon of HTRS and describes the range of physiological and psychosocial stressors that can produce a sensitized hypertensive state. Also discussed are the cellular and molecular changes in the brain neural network controlling sympathetic tone involved in long-term storage of information relating to stressors, which could serve to maintain a sensitized state. Finally, this Review concludes with a discussion of why a sensitized hypertensive response might previously have been beneficial and increased biological fitness under some environmental conditions and why today it has become a health-related liability.
Collapse
Affiliation(s)
- Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA.
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, USA.
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA.
- The François M. Abboud Cardiovascular Center, Iowa City, IA, USA.
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- The François M. Abboud Cardiovascular Center, Iowa City, IA, USA
| |
Collapse
|
142
|
Korim WS, Elsaafien K, Basser JR, Setiadi A, May CN, Yao ST. In renovascular hypertension, TNF-α type-1 receptors in the area postrema mediate increases in cardiac and renal sympathetic nerve activity and blood pressure. Cardiovasc Res 2018; 115:1092-1101. [DOI: 10.1093/cvr/cvy268] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/02/2018] [Accepted: 10/24/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Willian S Korim
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Wade Institute of Entrepreneurship, University of Melbourne, Parkville, VIC, Australia
| | - Khalid Elsaafien
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Jeremy R Basser
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Anthony Setiadi
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Clive N May
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Song T Yao
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
143
|
Blockade of Endogenous Angiotensin-(1-7) in Hypothalamic Paraventricular Nucleus Attenuates High Salt-Induced Sympathoexcitation and Hypertension. Neurosci Bull 2018; 35:47-56. [PMID: 30328008 DOI: 10.1007/s12264-018-0297-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 08/24/2018] [Indexed: 12/18/2022] Open
Abstract
Angiotensin (Ang)-(1-7) is an important biologically-active peptide of the renin-angiotensin system. This study was designed to determine whether inhibition of Ang-(1-7) in the hypothalamic paraventricular nucleus (PVN) attenuates sympathetic activity and elevates blood pressure by modulating pro-inflammatory cytokines (PICs) and oxidative stress in the PVN in salt-induced hypertension. Rats were fed either a high-salt (8% NaCl) or a normal salt diet (0.3% NaCl) for 10 weeks, followed by bilateral microinjections of the Ang-(1-7) antagonist A-779 or vehicle into the PVN. We found that the mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA), and plasma norepinephrine (NE) were significantly increased in salt-induced hypertensive rats. The high-salt diet also resulted in higher levels of the PICs interleukin-6, interleukin-1beta, tumor necrosis factor alpha, and monocyte chemotactic protein-1, as well as higher gp91phox expression and superoxide production in the PVN. Microinjection of A-779 (3 nmol/50 nL) into the bilateral PVN of hypertensive rats not only attenuated MAP, RSNA, and NE, but also decreased the PICs and oxidative stress in the PVN. These results suggest that the increased MAP and sympathetic activity in salt-induced hypertension can be suppressed by blockade of endogenous Ang-(1-7) in the PVN, through modulation of PICs and oxidative stress.
Collapse
|
144
|
Maglione AV, Taranto P, Hamermesz B, Souza JS, Cafarchio EM, Ogihara CA, Maciel RMB, Giannocco G, Sato MA. Impact of swimming exercise on inflammation in medullary areas of sympathetic outflow control in spontaneously hypertensive rats. Metab Brain Dis 2018; 33:1649-1660. [PMID: 29946957 DOI: 10.1007/s11011-018-0273-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Exercise reduces sympathetic activity (SA), arterial pressure and heart rate in spontaneously hypertensive rats (SHR). Exercise increases oxidative stress (OS) and inflammation is implicated in the generation of reactive oxygen species (ROS) and progression of hypertension. To unravel these effects of exercise and considering that SA is driven by medullary areas, we hypothesized that swimming exercise (SW) affects the gene expression (g.e.) of proteins involved in inflammation and OS in the commissural Nucleus of the Solitary Tract (cNTS) and Rostral ventrolateral medulla (RVLM), which control the sympathetic outflow in SHR. We used male SHR and Wistar rats (14-16wks-old) which were maintained sedentary (SED) or submitted to SW (1 h/day, 5 days/wk./6wks). The g.e. of cycloxygenase-2 (COX-2), interleukin 6 (IL-6), interleukin 10 (IL-10), AT-1 receptor (AT-1r), neuroglobin (Ngb) and cytoglobin (Ctb) in cNTS and RVLM was carried out by qPCR. We observed that COX-2 g.e. increased in SW-SHR in cNTS and RVLM compared to SED-SHR. The IL-6 g.e. reduced in RVLM in SW-SHR, whereas IL-10 g.e. increased in SW-SHR in comparison to SED-SHR. The AT-1r g.e. decreased in SW-SHR in cNTS and RVLM compared to SED-SHR. The Ngb and Ctb g.e. in cNTS neurons increased in SHR and Wistar rats submitted to SW compared to SED, but only Ctb g.e. increased in RVLM in SW-SHR and Wistar in comparison to SED. Therefore, the SW altered the g.e. in cNTS and RVLM for reducing the inflammation and ROS formation, which is increased particularly in SHR, consequently decreasing the OS.
Collapse
Affiliation(s)
- Andrea V Maglione
- Department of Morphology and Physiology, Faculdade de Medicina do ABC, 2000 Lauro Gomes Ave., Vila Sacadura Cabral, Santo Andre, SP, 09060-870, Brazil
| | - Patrícia Taranto
- Department of Morphology and Physiology, Faculdade de Medicina do ABC, 2000 Lauro Gomes Ave., Vila Sacadura Cabral, Santo Andre, SP, 09060-870, Brazil
| | - Bruno Hamermesz
- Department of Morphology and Physiology, Faculdade de Medicina do ABC, 2000 Lauro Gomes Ave., Vila Sacadura Cabral, Santo Andre, SP, 09060-870, Brazil
| | - Janaina S Souza
- Department of Medicine, Federal Univesity of Sao Paulo, Sao Paulo, SP, Brazil
| | - Eduardo M Cafarchio
- Department of Morphology and Physiology, Faculdade de Medicina do ABC, 2000 Lauro Gomes Ave., Vila Sacadura Cabral, Santo Andre, SP, 09060-870, Brazil
| | - Cristiana A Ogihara
- Department of Morphology and Physiology, Faculdade de Medicina do ABC, 2000 Lauro Gomes Ave., Vila Sacadura Cabral, Santo Andre, SP, 09060-870, Brazil
| | - Rui M B Maciel
- Department of Medicine, Federal Univesity of Sao Paulo, Sao Paulo, SP, Brazil
| | - Gisele Giannocco
- Department of Medicine, Federal Univesity of Sao Paulo, Sao Paulo, SP, Brazil
| | - Monica A Sato
- Department of Morphology and Physiology, Faculdade de Medicina do ABC, 2000 Lauro Gomes Ave., Vila Sacadura Cabral, Santo Andre, SP, 09060-870, Brazil.
| |
Collapse
|
145
|
Mi Y, Wu Q, Yuan W, Chen F, Du D. Role of microglia M1/M2 polarisation in the paraventricular nucleus: New insight into the development of stress-induced hypertension in rats. Auton Neurosci 2018; 213:71-80. [DOI: 10.1016/j.autneu.2018.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
|
146
|
Cohen EM, Farnham MMJ, Kakall Z, Kim SJ, Nedoboy PE, Pilowsky PM. Glia and central cardiorespiratory pathology. Auton Neurosci 2018; 214:24-34. [PMID: 30172674 DOI: 10.1016/j.autneu.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 01/08/2023]
Abstract
Respiration and blood pressure are primarily controlled by somatic and autonomic motor neurones, respectively. Central cardiorespiratory control is critical in moment-to-moment survival, but it also has a role in the development and maintenance of chronic pathological conditions such as hypertension. The glial cells of the brain are non-neuronal cells with metabolic, immune, and developmental functions. Recent evidence shows that glia play an active role in supporting and regulating the neuronal circuitry which drives the cardiorespiratory system. Here we will review the activities of two key types of glial cell, microglia and astrocytes, in assisting normal central cardiorespiratory control and in pathology.
Collapse
Affiliation(s)
- E Myfanwy Cohen
- The Heart Research Institute, Sydney, New South Wales 2042, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Melissa M J Farnham
- The Heart Research Institute, Sydney, New South Wales 2042, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zohra Kakall
- The Heart Research Institute, Sydney, New South Wales 2042, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Seung Jae Kim
- The Heart Research Institute, Sydney, New South Wales 2042, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Polina E Nedoboy
- The Heart Research Institute, Sydney, New South Wales 2042, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Paul M Pilowsky
- The Heart Research Institute, Sydney, New South Wales 2042, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
147
|
Yamazato M, Ishida A, Yamazato Y, Nakamura T, Ohya Y. Intracerebroventricular administration of bone marrow-derived cells attenuates angiotensin II-initiated neurogenic hypertension in rats. Hypertens Res 2018; 41:828-838. [DOI: 10.1038/s41440-018-0088-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 11/09/2022]
|
148
|
Abstract
PURPOSE OF REVIEW Inflammatory cytokines contribute to the pathogenesis of hypertension through effects on renal blood flow and sodium handling. This review will update recent advances that explore the renal actions of immune cells and cytokines in the pathogenesis of hypertension. RECENT FINDINGS Populations of cells from both the innate and adaptive immune systems contribute to hypertension by modulating functions of the vasculature and epithelial cells in the kidney. Macrophages and T lymphocytes can directly regulate the hypertensive response and consequent target organ damage. Dendritic cells and B lymphocytes can alter blood pressure (BP) indirectly by facilitating T-cell activation. Proinflammatory cytokines, including tumor necrosis factor-α, interleukin 17, interleukin 1, and interferon-γ augment BP and/or renal injury when produced by T helper 1 cells, T helper 17 cells, and macrophages. In contrast, interleukin 10 improves vascular and renal functions in preclinical hypertension studies. The effects of transforming growth factor-β are complex because of its profibrotic and immunosuppressive functions that also depend on the localization and concentration of this pleiotropic cytokine. SUMMARY Preclinical studies point to a key role for cytokines in hypertension via their actions in the kidney. Consistent with this notion, anti-inflammatory therapies can attenuate BP elevation in human patients with rheumatologic disease. Conversely, impaired natriuresis may further polarize both T lymphocytes and macrophages toward a proinflammatory state, in a pathogenic, feed-forward loop of immune activation and BP elevation. Understanding the precise renal actions of cytokines in hypertension will be necessary to inhibit cytokine-dependent hypertensive responses while preserving systemic immunity and tumor surveillance.
Collapse
|
149
|
Nunes KP, de Oliveira AA, Mowry FE, Biancardi VC. Targeting toll-like receptor 4 signalling pathways: can therapeutics pay the toll for hypertension? Br J Pharmacol 2018; 176:1864-1879. [PMID: 29981161 DOI: 10.1111/bph.14438] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/09/2018] [Accepted: 06/24/2018] [Indexed: 02/06/2023] Open
Abstract
The immune system plays a prominent role in the initiation and maintenance of hypertension. The innate immune system, via toll-like receptors (TLRs), identifies distinct signatures of invading microbes and damage-associated molecular patterns and triggers a chain of downstream signalling cascades, leading to secretion of pro-inflammatory cytokines and shaping the adaptive immune response. Over the past decade, a dysfunctional TLR-mediated response, particularly via TLR4, has been suggested to support a chronic inflammatory state in hypertension, inducing deleterious local and systemic effects in host cells and tissues and contributing to disease progression. While the underlying mechanisms triggering TLR4 need further research, evidence suggests that sustained elevations in BP disrupt homeostasis, releasing endogenous TLR4 ligands in hypertension. In this review, we discuss the emerging role of TLR4 in the pathogenesis of hypertension and whether targeting this receptor and its signalling pathways could offer a therapeutic strategy for management of this multifaceted disease. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Kenia Pedrosa Nunes
- Department of Biological Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | | | | | | |
Collapse
|
150
|
Haspula D, Clark MA. Molecular Basis of the Brain Renin Angiotensin System in Cardiovascular and Neurologic Disorders: Uncovering a Key Role for the Astroglial Angiotensin Type 1 Receptor AT1R. J Pharmacol Exp Ther 2018; 366:251-264. [PMID: 29752427 DOI: 10.1124/jpet.118.248831] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
The central renin angiotensin system (RAS) is one of the most widely investigated cardiovascular systems in the brain. It is implicated in a myriad of cardiovascular diseases. However, studies from the last decade have identified its involvement in several neurologic abnormalities. Understanding the molecular functionality of the various RAS components can thus provide considerable insight into the phenotypic differences and mechanistic drivers of not just cardiovascular but also neurologic disorders. Since activation of one of its primary receptors, the angiotensin type 1 receptor (AT1R), results in an augmentation of oxidative stress and inflammatory cytokines, it becomes essential to investigate not just neuronal RAS but glial RAS as well. Glial cells are key homeostatic regulators in the brain and are critical players in the resolution of overt oxidative stress and neuroinflammation. Designing better and effective therapeutic strategies that target the brain RAS could well hinge on understanding the molecular basis of both neuronal and glial RAS. This review provides a comprehensive overview of the major studies that have investigated the mechanisms and regulation of the brain RAS, and it also provides insight into the potential role of glial AT1Rs in the pathophysiology of cardiovascular and neurologic disorders.
Collapse
Affiliation(s)
- Dhanush Haspula
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin (D.H.); and College of Pharmacy, Department of Pharmaceutical Sciences, Nova Southeastern University, Ft. Lauderdale, Florida (M.A.C.)
| | - Michelle A Clark
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin (D.H.); and College of Pharmacy, Department of Pharmaceutical Sciences, Nova Southeastern University, Ft. Lauderdale, Florida (M.A.C.)
| |
Collapse
|