151
|
Patrick DM, Van Beusecum JP, Kirabo A. The role of inflammation in hypertension: novel concepts. CURRENT OPINION IN PHYSIOLOGY 2020; 19:92-98. [PMID: 33073072 PMCID: PMC7552986 DOI: 10.1016/j.cophys.2020.09.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hypertension remains the most important modifiable risk factor for the development of cardiovascular disease. While it is clear that inflammation plays a pivotal role in the development and maintenance of hypertension, several novel discoveries have been made within the past decade that have advanced the field and have provided new mechanistic insights. First, recent studies have identified a central role of sodium-induced immune cell activation in the pathogenesis of hypertension by altering the gut microbiome and formation of products of lipid oxidation known as isolevuglandins. Second, cytokine elaboration by the inflammasome leading to end-organ dysfunction and immune activation has been found to play a role in the genesis of hypertension. Third, novel techniques have identified previously uncharacterized immune cell populations that may play a functional role in these processes. Finally, the role of inflammation in hypertension may be an important mediator of severe COVID-19 infections. In this review, we discuss these recent advances in the study of inflammation and hypertension and highlight topics for future studies.
Collapse
Affiliation(s)
- David M Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, USA
| | - Justin P Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
152
|
Macrophage secretion of miR-106b-5p causes renin-dependent hypertension. Nat Commun 2020; 11:4798. [PMID: 32968066 PMCID: PMC7511948 DOI: 10.1038/s41467-020-18538-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/24/2020] [Indexed: 12/28/2022] Open
Abstract
Myeloid cells are known mediators of hypertension, but their role in initiating renin-induced hypertension has not been studied. Vitamin D deficiency causes pro-inflammatory macrophage infiltration in metabolic tissues and is linked to renin-mediated hypertension. We tested the hypothesis that impaired vitamin D signaling in macrophages causes hypertension using conditional knockout of the myeloid vitamin D receptor in mice (KODMAC). These mice develop renin-dependent hypertension due to macrophage infiltration of the vasculature and direct activation of renal juxtaglomerular (JG) cell renin production. Induction of endoplasmic reticulum stress in knockout macrophages increases miR-106b-5p secretion, which stimulates JG cell renin production via repression of transcription factors E2f1 and Pde3b. Moreover, in wild-type recipient mice of KODMAC/miR106b−/− bone marrow, knockout of miR-106b-5p prevents the hypertension and JG cell renin production induced by KODMAC macrophages, suggesting myeloid-specific, miR-106b-5p-dependent effects. These findings confirm macrophage miR-106b-5p secretion from impaired vitamin D receptor signaling causes inflammation-induced hypertension. Myeloid cells are involved in hypertension, but their exact role in renin-induced hypertension remains unclear. Here the authors show that impaired vitamin D signaling in myeloid cells causes hypertension via macrophage-specific miR-106b-5p secretion, which activates renin production in the kidney.
Collapse
|
153
|
Influenza A virus causes maternal and fetal pathology via innate and adaptive vascular inflammation in mice. Proc Natl Acad Sci U S A 2020; 117:24964-24973. [PMID: 32958663 PMCID: PMC7547222 DOI: 10.1073/pnas.2006905117] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Influenza infection during pregnancy is associated with increased maternal and perinatal complications. Here, we show that, during pregnancy, influenza infection leads to viral dissemination into the aorta, resulting in a peripheral “vascular storm” characterized by enhanced inflammatory mediators; the influx of Ly6C monocytes, neutrophils, and T cells; and impaired vascular function. The ensuing vascular storm induced hypoxia in the placenta and fetal brain and caused an increase in circulating cell free fetal DNA and soluble Flt1 release. We demonstrate that vascular dysfunction occurs in response to viral infection during pregnancy, which may explain the high rates of morbidity and mortality in pregnant dams, as well as the downstream perinatal complications associated with influenza infection. Influenza A virus (IAV) infection during pregnancy causes severe maternal and perinatal complications, despite a lack of vertical transmission of IAV across the placenta. Here, we demonstrate a significant alteration in the maternal vascular landscape that underpins the maternal and downstream fetal pathology to IAV infection in mice. In IAV infection of nonpregnant mice, the local lung inflammatory response was contained to the lungs and was self-resolving, whereas in pregnant mice, virus dissemination to major maternal blood vessels, including the aorta, resulted in a peripheral "vascular storm," with elevated proinflammatory and antiviral mediators and the influx of Ly6Clow and Ly6Chigh monocytes, plus neutrophils and T cells. This vascular storm was associated with elevated levels of the adhesion molecules ICAM and VCAM and the pattern-recognition receptors TLR7 and TLR9 in the vascular wall, resulting in profound vascular dysfunction. The sequalae of this IAV-driven vascular storm included placental growth retardation and intrauterine growth restriction, evidence of placental and fetal brain hypoxia, and increased circulating cell free fetal DNA and soluble Flt1. In contrast, IAV infection in nonpregnant mice caused no obvious alterations in endothelial function or vascular inflammation. Therefore, IAV infection during pregnancy drives a significant systemic vascular alteration in pregnant dams, which likely suppresses critical blood flow to the placenta and fetus. This study in mice provides a fundamental mechanistic insight and a paradigm into how an immune response to a respiratory virus, such as IAV, is likely to specifically drive maternal and fetal pathologies during pregnancy.
Collapse
|
154
|
He X, Deng J, Yu XJ, Yang S, Yang Y, Zang WJ. Activation of M3AChR (Type 3 Muscarinic Acetylcholine Receptor) and Nrf2 (Nuclear Factor Erythroid 2-Related Factor 2) Signaling by Choline Alleviates Vascular Smooth Muscle Cell Phenotypic Switching and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2020; 40:2649-2664. [PMID: 32938216 DOI: 10.1161/atvbaha.120.315146] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Phenotypic switching of vascular smooth muscle cells (VSMCs) plays a critical role in atherosclerosis, vascular restenosis, and hypertension. Choline exerts cardioprotective effects; however, little is known about its effects on VSMC phenotypic switching and vascular remodeling. Here, we investigated whether choline modulates VSMC phenotypic changes and explored the underlying mechanisms. Approach and Results: In cultured VSMCs, choline promoted Nrf2 (nuclear factor erythroid 2-related factor 2) nuclear translocation, inducing the expression of HO-1 (heme oxygenase-1) and NQO-1 (NAD[P]H quinone oxidoreductase-1). Consequently, choline ameliorated Ang II (angiotensin II)-induced increases in NOX (NAD[P]H oxidase) expression and the mitochondrial reactive oxygen species level, thereby attenuating Ang II-induced VSMC phenotypic switching, proliferation, and migration, presumably via M3AChRs (type 3 muscarinic acetylcholine receptors). Downregulation of M3AChR or Nrf2 diminished choline-mediated upregulation of Nrf2, HO-1, and NQO-1 expression, as well as inhibition of VSMC phenotypic transformation, suggesting that M3AChR and Nrf2 activation are responsible for the protective effects of choline. Moreover, activation of the Nrf2 pathway by sulforaphane suppressed Ang II-induced VSMC phenotypic switching and proliferation, indicating that Nrf2 is a key regulator of VSMC phenotypic switching and vascular homeostasis. In a rat model of abdominal aortic constriction in vivo, choline attenuated VSMC phenotypic transformation and vascular remodeling in a manner related to activation of the Nrf2 pathway. CONCLUSIONS These results reveal that choline impedes VSMC phenotypic switching, proliferation, migration, and vascular remodeling by activating M3AChR and Nrf2-antioxidant signaling and suggest a novel role for Nrf2 in VSMC phenotypic modulation.
Collapse
Affiliation(s)
- Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China
| | - Juan Deng
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China
| | - Xiao-Jiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China
| | - Si Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China
| | - Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China
| |
Collapse
|
155
|
Thomas TP, Grisanti LA. The Dynamic Interplay Between Cardiac Inflammation and Fibrosis. Front Physiol 2020; 11:529075. [PMID: 33041853 PMCID: PMC7522448 DOI: 10.3389/fphys.2020.529075] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Heart failure is a leading cause of death worldwide. While there are multiple etiologies contributing to the development of heart failure, all cause result in impairments in cardiac function that is characterized by changes in cardiac remodeling and compliance. Fibrosis is associated with nearly all forms of heart failure and is an important contributor to disease pathogenesis. Inflammation also plays a critical role in the heart and there is a large degree of interconnectedness between the inflammatory and fibrotic response. This review discusses the cellular and molecular mechanisms contributing to inflammation and fibrosis and the interplay between the two.
Collapse
Affiliation(s)
- Toby P Thomas
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
156
|
Weinberger T, Esfandyari D, Messerer D, Percin G, Schleifer C, Thaler R, Liu L, Stremmel C, Schneider V, Vagnozzi RJ, Schwanenkamp J, Fischer M, Busch K, Klapproth K, Ishikawa-Ankerhold H, Klösges L, Titova A, Molkentin JD, Kobayashi Y, Engelhardt S, Massberg S, Waskow C, Perdiguero EG, Schulz C. Ontogeny of arterial macrophages defines their functions in homeostasis and inflammation. Nat Commun 2020; 11:4549. [PMID: 32917889 PMCID: PMC7486394 DOI: 10.1038/s41467-020-18287-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
Arterial macrophages have different developmental origins, but the association of macrophage ontogeny with their phenotypes and functions in adulthood is still unclear. Here, we combine macrophage fate-mapping analysis with single-cell RNA sequencing to establish their cellular identity during homeostasis, and in response to angiotensin-II (AngII)-induced arterial inflammation. Yolk sac erythro-myeloid progenitors (EMP) contribute substantially to adventitial macrophages and give rise to a defined cluster of resident immune cells with homeostatic functions that is stable in adult mice, but declines in numbers during ageing and is not replenished by bone marrow (BM)-derived macrophages. In response to AngII inflammation, increase in adventitial macrophages is driven by recruitment of BM monocytes, while EMP-derived macrophages proliferate locally and provide a distinct transcriptional response that is linked to tissue regeneration. Our findings thus contribute to the understanding of macrophage heterogeneity, and associate macrophage ontogeny with distinct functions in health and disease. Arterial macrophages develop from either yolk sac or bone marrow progenitors. Here, the author show that ageing-induced reduction of arterial macrophages is not replenished by bone marrow-derived cells, but under inflammatory conditions circulating monocytes are recruited to maintain homeostasis, while arterial macrophages of yolk sac origin carry out tissue repair.
Collapse
Affiliation(s)
- Tobias Weinberger
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Dena Esfandyari
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany.,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Strasse 29, 80802, Munich, Germany
| | - Denise Messerer
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Gulce Percin
- Regeneration in Hematopoiesis, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Christian Schleifer
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Raffael Thaler
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Lulu Liu
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Christopher Stremmel
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Vanessa Schneider
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Ronald J Vagnozzi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Jennifer Schwanenkamp
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Maximilian Fischer
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Kay Klapproth
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Hellen Ishikawa-Ankerhold
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Lukas Klösges
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Anna Titova
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yasuhiro Kobayashi
- Institute for Oral Science, Matsumoto Dental University, 1780 Hiro-Oka Gobara Shiojiri, Nagano, 390-0781, Japan
| | - Stefan Engelhardt
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany.,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Strasse 29, 80802, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany.,Faculty of Biological Sciences, Friedrich-Schiller-University Jena, 07737 Jena, 07745, Jena, Germany
| | - Elisa Gomez Perdiguero
- Institut Pasteur, Macrophages and Endothelial cells, Département de Biologie du Développement et Cellules Souches, UMR3738 CNRS, Paris, 75015, France
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany. .,Walter-Brendel-Center for Experimental Medicine, Ludwig Maximilian University, Marchioninistrasse 27, 81377, Munich, Germany.
| |
Collapse
|
157
|
Czopek A, Moorhouse R, Guyonnet L, Farrah T, Lenoir O, Owen E, van Bragt J, Costello HM, Menolascina F, Baudrie V, Webb DJ, Kluth DC, Bailey MA, Tharaux PL, Dhaun N. A novel role for myeloid endothelin-B receptors in hypertension. Eur Heart J 2020; 40:768-784. [PMID: 30657897 PMCID: PMC6396028 DOI: 10.1093/eurheartj/ehy881] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/04/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022] Open
Abstract
AIMS Hypertension is common. Recent data suggest that macrophages (Mφ) contribute to, and protect from, hypertension. Endothelin-1 (ET-1) is the most potent endogenous vasoconstrictor with additional pro-inflammatory properties. We investigated the role of the ET system in experimental and clinical hypertension by modifying Mφ number and phenotype. METHODS AND RESULTS In vitro, Mφ ET receptor function was explored using pharmacological, gene silencing, and knockout approaches. Using the CD11b-DTR mouse and novel mice with myeloid cell-specific endothelin-B (ETB) receptor deficiency (LysMETB-/-), we explored the effects of modifying Mφ number and phenotype on the hypertensive effects of ET-1, angiotensin II (ANG II), a model that is ET-1 dependent, and salt. In patients with small vessel vasculitis, the impacts of Mφ depleting and non-depleting therapies on blood pressure (BP) and endothelial function were examined. Mouse and human Mφ expressed both endothelin-A and ETB receptors and displayed chemokinesis to ET-1. However, stimulation of Mφ with exogenous ET-1 did not polarize Mφ phenotype. Interestingly, both mouse and human Mφ cleared ET-1 through ETB receptor mediated, and dynamin-dependent, endocytosis. Mφ depletion resulted in an augmented chronic hypertensive response to both ET-1 and salt. LysMETB-/- mice displayed an exaggerated hypertensive response to both ET-1 and ANG II. Finally, in patients who received Mφ depleting immunotherapy BP was higher and endothelial function worse than in those receiving non-depleting therapies. CONCLUSION Mφ and ET-1 may play an important role in BP control and potentially have a critical role as a therapeutic target in hypertension.
Collapse
Affiliation(s)
- Alicja Czopek
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Rebecca Moorhouse
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Léa Guyonnet
- Paris Cardiovascular Research Centre - PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Tariq Farrah
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Olivia Lenoir
- Paris Cardiovascular Research Centre - PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Elizabeth Owen
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Job van Bragt
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Hannah M Costello
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Filippo Menolascina
- School of Engineering & SynthSys, Institute for Bioengineering, Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh, UK.,MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Véronique Baudrie
- Paris Cardiovascular Research Centre - PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - David J Webb
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - David C Kluth
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Matthew A Bailey
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Centre - PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Neeraj Dhaun
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK.,Paris Cardiovascular Research Centre - PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
158
|
Neural Control of Immunity in Hypertension: Council on Hypertension Mid Career Award for Research Excellence, 2019. Hypertension 2020; 76:622-628. [DOI: 10.1161/hypertensionaha.120.14637] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The nervous system and the immune system share the common ability to exert gatekeeper roles at the interfaces between internal and external environment. Although interaction between these 2 evolutionarily highly conserved systems has been recognized for long time, the investigation into the pathophysiological mechanisms underlying their crosstalk has been tackled only in recent decades. Recent work of the past years elucidated how the autonomic nervous system controls the splenic immunity recruited by hypertensive challenges. This review will focus on the neural mechanisms regulating the immune response and the role of this neuroimmune crosstalk in hypertension. In this context, the review highlights the components of the brain-spleen axis with a focus on the neuroimmune interface established in the spleen, where neural signals shape the immune response recruited to target organs of high blood pressure.
Collapse
|
159
|
Daiber A, Chlopicki S. Revisiting pharmacology of oxidative stress and endothelial dysfunction in cardiovascular disease: Evidence for redox-based therapies. Free Radic Biol Med 2020; 157:15-37. [PMID: 32131026 DOI: 10.1016/j.freeradbiomed.2020.02.026] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/05/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
According to the latest Global Burden of Disease Study data, non-communicable diseases in general and cardiovascular disease (CVD) in particular are the leading cause of premature death and reduced quality of life. Demographic shifts, unhealthy lifestyles and a higher burden of adverse environmental factors provide an explanation for these findings. The expected growing prevalence of CVD requires enhanced research efforts for identification and characterisation of novel therapeutic targets and strategies. Cardiovascular risk factors including classical (e.g. hypertension, diabetes, hypercholesterolaemia) and non-classical (e.g. environmental stress) factors induce the development of endothelial dysfunction, which is closely associated with oxidant stress and vascular inflammation and results in CVD, particularly in older adults. Most classically successful therapies for CVD display vasoprotective, antioxidant and anti-inflammatory effects, but were originally designed with other therapeutic aims. So far, only a few 'redox drugs' are in clinical use and many antioxidant strategies have not met expectations. With the present review, we summarise the actual knowledge on CVD pathomechanisms, with special emphasis on endothelial dysfunction, adverse redox signalling and oxidative stress, highlighting the preclinical and clinical evidence. In addition, we provide a brief overview of established CVD therapies and their relation to endothelial dysfunction and oxidative stress. Finally, we discuss novel strategies for redox-based CVD therapies trying to explain why, despite a clear link between endothelial dysfunction and adverse redox signalling and oxidative stress, redox- and oxidative stress-based therapies have not yet provided a breakthrough in the treatment of endothelial dysfunction and CVD.
Collapse
Affiliation(s)
- Andreas Daiber
- The Center for Cardiology, Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; The Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Stefan Chlopicki
- The Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland; Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
160
|
Abstract
PURPOSE OF REVIEW The main goal of this article is to discuss the role of the epithelial sodium channel (ENaC) in extracellular fluid and blood pressure regulation. RECENT FINDINGS Besides its role in sodium handling in the kidney, recent studies have found that ENaC expressed in other cells including immune cells can influence blood pressure via extra-renal mechanisms. Dendritic cells (DCs) are activated and contribute to salt-sensitive hypertension in an ENaC-dependent manner. We discuss recent studies on how ENaC is regulated in both the kidney and other sites including the vascular smooth muscles, endothelial cells, and immune cells. We also discuss how this extra-renal ENaC can play a role in salt-sensitive hypertension and its promise as a novel therapeutic target. The role of ENaC in blood pressure regulation in the kidney has been well studied. Recent human gene sequencing efforts have identified thousands of variants among the genes encoding ENaC, and research efforts to determine if these variants and their expression in extra-renal tissue play a role in hypertension will advance our understanding of the pathogenesis of ENaC-mediated cardiovascular disease and lead to novel therapeutic targets.
Collapse
Affiliation(s)
- Ashley L Pitzer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN, 37232, USA
| | - Justin P Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN, 37232, USA
| | - Thomas R Kleyman
- Departments of Medicine, Cell Biology, Pharmacology, and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN, 37232, USA. .,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
161
|
Targeted depletion of monocyte/macrophage suppresses aortic dissection with the spatial regulation of MMP-9 in the aorta. Life Sci 2020; 254:116927. [DOI: 10.1016/j.lfs.2019.116927] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022]
|
162
|
Abstract
PURPOSE OF REVIEW Inflammatory processes play a critical role in the pathogenesis of hypertension. Innate and adaptive immune responses participate in blood pressure (BP) elevation and end-organ damage. In this review, we discuss recent studies illustrating mechanisms through which immune cells and cytokines regulate BP via their actions in the kidney. RECENT FINDINGS Cells of the innate immune system, including monocytes, neutrophils, and dendritic cells, can all promote BP elevation via effects on kidney function. These innate immune cells can directly impact oxidative stress and cytokine generation in the kidney and/or present antigens to lymphocytes for the engagement of the adaptive immune system. Once activated by dendritic cells, effector memory T cells accumulate in the hypertensive kidney and facilitate renal salt and water retention. Individual subsets of activated T cells can secrete tumor necrosis factor-alpha (TNF-α), interleukin-17a (IL-17a), and interferon-gamma (IFN-γ), each of which has augmented the elevation of blood pressure in hypertensive models by enhancing renal sodium transport. B cells, regulate blood pressure via vasopressin receptor 2 (V2R)-dependent effects on fluid transport in the kidney. SUMMARY Immune cells of the innate and adaptive immune systems drive sodium retention and blood pressure elevation in part by altering renal solute transport.
Collapse
|
163
|
Li H, Xia N. The role of oxidative stress in cardiovascular disease caused by social isolation and loneliness. Redox Biol 2020; 37:101585. [PMID: 32709420 PMCID: PMC7767744 DOI: 10.1016/j.redox.2020.101585] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Loneliness and social isolation are common sources of chronic stress in modern society. Epidemiological studies have demonstrated that loneliness and social isolation increase mortality risk as much as smoking or alcohol consumption and more than physical inactivity or obesity. Loneliness in human is associated with higher blood pressure whereas enhanced atherosclerosis is observed in animal models of social isolation. Loneliness and social isolation lead to activation of the hypothalamic-pituitary-adrenocortical (HPA) axis, enhanced sympathetic nerve activity, impaired parasympathetic function and a proinflammatory immune response. These mechanisms have been implicated in the development of cardiovascular disease conferred by social isolation although a causal relationship has not been established so far. There is evidence that oxidative stress is likely to be a key molecular mechanism linking chronic psychosocial stress to cardiovascular disease. NADPH oxidase-mediated oxidative stress in the hypothalamus has been shown to be required for social isolation-induced HPA axis activation in socially isolated rats. Oxidative stress in the rostral ventrolateral medulla is also a key regulator of sympathetic nerve activity. In the vasculature, oxidative stress increases vascular tone and promote atherogenesis through multiple mechanisms. Thus, preventing oxidative stress may represent a therapeutic strategy to reduce the detrimental effects of social stress on health.
Collapse
Affiliation(s)
- Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131, Mainz, Germany.
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131, Mainz, Germany.
| |
Collapse
|
164
|
Angiotensin II Infusion Leads to Aortic Dissection in LRP8 Deficient Mice. Int J Mol Sci 2020; 21:ijms21144916. [PMID: 32664652 PMCID: PMC7404218 DOI: 10.3390/ijms21144916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 01/27/2023] Open
Abstract
Myeloid cells are crucial for the development of vascular inflammation. Low-density lipoprotein receptor-related protein 8 (LRP8) or Apolipoprotein E receptor 2 (ApoER2), is expressed by macrophages, endothelial cells and platelets and has been implicated in the development of cardiovascular diseases. Our aim was to evaluate the role of LRP8, in particular from immune cells, in the development of vascular inflammation. Methods. LRP8+/+ and LRP8−/− mice (on B6;129S background) were infused with angiotensin II (AngII, 1 mg/kg/day for 7 to 28 day) using osmotic minipumps. Blood pressure was recorded using tail cuff measurements. Vascular reactivity was assessed in isolated aortic segments. Leukocyte activation and infiltration were assessed by flow cytometry of aortic tissue and intravital videomicroscopy imaging. Histological analysis of aortic sections was conducted using sirius red staining. Results. AngII infusion worsened endothelial-dependent vascular relaxation and immune cells rolling and adherence to the carotid artery in both LRP8+/+ as well as LRP8−/− mice. However, only LRP8−/− mice demonstrated a drastically increased mortality rate in response to AngII due to aortic dissection. Bone marrow transplantation revealed that chimeras with LRP8 deficient myeloid cells phenocopied LRP8−/− mice. Conclusion. AngII-infused LRP8 deficient mice could be a useful animal model to study aortic dissection reflecting the lethality of this disease in humans.
Collapse
|
165
|
Dendritic cells are crucial for cardiovascular remodeling and modulate neutrophil gelatinase-associated lipocalin expression upon mineralocorticoid receptor activation. J Hypertens 2020; 37:1482-1492. [PMID: 31033725 DOI: 10.1097/hjh.0000000000002067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Adaptive immunity is crucial in cardiovascular and renal inflammation/fibrosis upon hyperactivation of mineralocorticoid receptor. We have previously demonstrated that dendritic cells can respond to mineralocorticoid receptor activation, and the neutrophil gelatinase-associated lipocalin (NGAL) in dendritic cells is highly increased during aldosterone (Aldo)/mineralocorticoid receptor-dependent cardiovascular damage. However, the interrelationship among dendritic cells, target organs inflammation/fibrosis induced by mineralocorticoid receptor, and NGAL-dependence remains unknown. OBJECTIVE We studied the role of dendritic cells in mineralocorticoid receptor-dependent tissue remodeling and whether NGAL can modulate the inflammatory response of dendritic cells after mineralocorticoid receptor activation. METHODS Cardiovascular and renal remodeling induced by Aldo and high-salt diet [nephrectomy-Aldo-salt (NAS) model] were analyzed in CD11c.DOG mice, a model which allows dendritic cells ablation by using diphtheria toxin. In addition, in-vitro studies in NGAL-knock out dendritic cells were performed to determine the immunomodulatory role of NGAL upon Aldo treatment. RESULTS The ablation of dendritic cells prevented the development of cardiac hypertrophy, perivascular fibrosis, and the overexpression of NGAL, brain natriuretic peptide, and two profibrotic factors induced by NAS: collagen 1A1 and connective tissue growth factor. We determined that dendritic cells were not required to prevent renal hypertrophy/fibrosis induced by NAS. Between different immune cells analyzed, we observed that NGAL abundance was higher in antigen-presenting cells, while in-vitro studies showed that mineralocorticoid receptor stimulation in dendritic cells favored NGAL and IL-23 expression (p19 and p40 subunits), which are involved in the development of fibrosis and the Th17-driven response, respectively. CONCLUSION NGAL produced by dendritic cells may play a pivotal role in the activation of adaptive immunity that leads to cardiovascular fibrosis during mineralocorticoids excess.
Collapse
|
166
|
Deletion of the myeloid endothelin-B receptor confers long-term protection from angiotensin II-mediated kidney, eye and vessel injury. Kidney Int 2020; 98:1193-1209. [PMID: 32569653 PMCID: PMC7652550 DOI: 10.1016/j.kint.2020.05.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/25/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
The endothelin system may be an important player in hypertensive end-organ injury as endothelin-1 increases blood pressure and is pro-inflammatory. The immune system is emerging as an important regulator of blood pressure and we have shown that the early hypertensive response to angiotensin-II infusion was amplified in mice deficient of myeloid endothelin-B (ETB) receptors (LysM-CreEdnrblox/lox). Hypothesizing that these mice would display enhanced organ injury, we gave angiotensin-II to LysM-CreEdnrblox/lox and littermate controls (Ednrblox/lox) for six weeks. Unexpectedly, LysM-CreEdnrblox/lox mice were significantly protected from organ injury, with less proteinuria, glomerulosclerosis and inflammation of the kidney compared to controls. In the eye, LysM-CreEdnrblox/lox mice had fewer retinal hemorrhages, less microglial activation and less vessel rarefaction. Cardiac remodeling and dysfunction were similar in both groups at week six but LysM-CreEdnrblox/lox mice had better endothelial function. Although blood pressure was initially higher in LysM-CreEdnrblox/lox mice, this was not sustained. A natriuretic switch at about two weeks, due to enhanced ETB signaling in the kidney, induced a hypertensive reversal. By week six, blood pressure was lower in LysM-CreEdnrblox/lox mice than in controls. At six weeks, macrophages from LysM-CreEdnrblox/lox mice were more anti-inflammatory and had greater phagocytic ability compared to the macrophages of Ednrblox/lox mice. Thus, myeloid cell ETB receptor signaling drives this injury both through amplifying hypertension and by inflammatory polarization of macrophages.
Collapse
|
167
|
Han X, Zhang YL, Fu TT, Li PB, Cong T, Li HH. Blockage of UCHL1 activity attenuates cardiac remodeling in spontaneously hypertensive rats. Hypertens Res 2020; 43:1089-1098. [DOI: 10.1038/s41440-020-0486-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/29/2020] [Accepted: 04/01/2020] [Indexed: 01/20/2023]
|
168
|
Schiffrin EL, Engert JC. Periodontitis and hypertension: causally linked by immune mechanisms. Eur Heart J 2020; 40:3471-3473. [PMID: 31589300 DOI: 10.1093/eurheartj/ehz729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Ernesto L Schiffrin
- Lady Davis Institute for Medical Research, and Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - James C Engert
- Department of Medicine, McGill University, Montreal, QC, Canada.,McGill University Health Centre Research Institute, Montreal, Canada
| |
Collapse
|
169
|
Arkorful MA, Noren Hooten N, Zhang Y, Hewitt AN, Barrientos Sanchez L, Evans MK, Dluzen DF. MicroRNA-1253 Regulation of WASF2 (WAVE2) and its Relevance to Racial Health Disparities. Genes (Basel) 2020; 11:E572. [PMID: 32443852 PMCID: PMC7288301 DOI: 10.3390/genes11050572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/13/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
The prevalence of hypertension among African Americans (AAs) in the US is among the highest of any demographic and affects over two-thirds of AA women. Previous data from our laboratory suggest substantial differential gene expression (DGE) of mRNAs and microRNAs (miRNAs) exists within peripheral blood mononuclear cells (PBMCs) isolated from AA and white women with or without hypertension. We hypothesized that DGE by race may contribute to racial differences in hypertension. In a reanalysis of our previous dataset, we found that the Wiskott-Aldrich syndrome protein Verprolin-homologous protein 2 (WASF2 (also known as WAVE2)) is differentially expressed in AA women with hypertension, along with several other members of the actin cytoskeleton signaling pathway that plays a role in cell shape and branching of actin filaments. We performed an in silico miRNA target prediction analysis that suggested miRNA miR-1253 regulates WASF2. Transfection of miR-1253 mimics into human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs) significantly repressed WASF2 mRNA and protein levels (p < 0.05), and a luciferase reporter assay confirmed that miR-1253 regulates the WASF2 3' UTR (p < 0.01). miR-1253 overexpression in HUVECs significantly increased HUVEC lamellipodia formation (p < 0.01), suggesting the miR-1253-WASF2 interaction may play a role in cell shape and actin cytoskeleton function. Together, we have identified novel roles for miR-1253 and WASF2 in a hypertension-related disparities context. This may ultimately lead to the discovery of additional actin-related genes which are important in the vascular-related complications of hypertension and influence the disproportionate susceptibility to hypertension among AAs in general and AA women in particular.
Collapse
Affiliation(s)
- Mercy A. Arkorful
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA;
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD 21224, USA;
| | - Amirah N. Hewitt
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Lori Barrientos Sanchez
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Douglas F. Dluzen
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA;
| |
Collapse
|
170
|
Regulation of Vascular Function and Inflammation via Cross Talk of Reactive Oxygen and Nitrogen Species from Mitochondria or NADPH Oxidase-Implications for Diabetes Progression. Int J Mol Sci 2020; 21:ijms21103405. [PMID: 32408480 PMCID: PMC7279344 DOI: 10.3390/ijms21103405] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a key role for the development of cardiovascular, metabolic, and neurodegenerative disease. This concept has been proven by using the approach of genetic deletion of reactive oxygen and nitrogen species (RONS) producing, pro-oxidant enzymes as well as by the overexpression of RONS detoxifying, antioxidant enzymes leading to an amelioration of the severity of diseases. Vice versa, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of RONS detoxifying enzymes. We have previously identified cross talk mechanisms between different sources of RONS, which can amplify the oxidative stress-mediated damage. Here, the pathways and potential mechanisms leading to this cross talk are analyzed in detail and highlighted by selected examples from the current literature and own data including hypoxia, angiotensin II (AT-II)-induced hypertension, nitrate tolerance, aging, and others. The general concept of redox-based activation of RONS sources via “kindling radicals” and enzyme-specific “redox switches” as well as the interaction with redox-sensitive inflammatory pathways are discussed. Here, we present evidence for the existence of such cross talk mechanisms in the setting of diabetes and critically assess their contribution to the severity of diabetic complications.
Collapse
|
171
|
Raven PB, Young BE, Fadel PJ. Arterial Baroreflex Resetting During Exercise in Humans: Underlying Signaling Mechanisms. Exerc Sport Sci Rev 2020; 47:129-141. [PMID: 30921029 DOI: 10.1249/jes.0000000000000190] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The arterial baroreflex (ABR) resets during exercise in an intensity-dependent manner to operate around a higher blood pressure with maintained sensitivity. This review provides a historical perspective of ABR resetting and the involvement of other neural reflexes in mediating exercise resetting. Furthermore, we discuss potential underlying signaling mechanisms that may contribute to exercise ABR resetting in physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Peter B Raven
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth
| | - Benjamin E Young
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX
| |
Collapse
|
172
|
Steven S, Frenis K, Kalinovic S, Kvandova M, Oelze M, Helmstädter J, Hahad O, Filippou K, Kus K, Trevisan C, Schlüter KD, Boengler K, Chlopicki S, Frauenknecht K, Schulz R, Sorensen M, Daiber A, Kröller-Schön S, Münzel T. Exacerbation of adverse cardiovascular effects of aircraft noise in an animal model of arterial hypertension. Redox Biol 2020; 34:101515. [PMID: 32345536 PMCID: PMC7327989 DOI: 10.1016/j.redox.2020.101515] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Arterial hypertension is the most important risk factor for the development of cardiovascular disease. Recently, aircraft noise has been shown to be associated with elevated blood pressure, endothelial dysfunction, and oxidative stress. Here, we investigated the potential exacerbated cardiovascular effects of aircraft noise in combination with experimental arterial hypertension. C57BL/6J mice were infused with 0.5 mg/kg/d of angiotensin II for 7 days, exposed to aircraft noise for 7 days at a maximum sound pressure level of 85 dB(A) and a mean sound pressure level of 72 dB(A), or subjected to both stressors. Noise and angiotensin II increased blood pressure, endothelial dysfunction, oxidative stress and inflammation in aortic, cardiac and/or cerebral tissues in single exposure models. In mice subjected to both stressors, most of these risk factors showed potentiated adverse changes. We also found that mice exposed to both noise and ATII had increased phagocytic NADPH oxidase (NOX-2)-mediated superoxide formation, immune cell infiltration (monocytes, neutrophils and T cells) in the aortic wall, astrocyte activation in the brain, enhanced cytokine signaling, and subsequent vascular and cerebral oxidative stress. Exaggerated renal stress response was also observed. In summary, our results show an enhanced adverse cardiovascular effect between environmental noise exposure and arterial hypertension, which is mainly triggered by vascular inflammation and oxidative stress. Mechanistically, noise potentiates neuroinflammation and cerebral oxidative stress, which may be a potential link between both risk factors. The results indicate that a combination of classical (arterial hypertension) and novel (noise exposure) risk factors may be deleterious for cardiovascular health. Noise exposure causes non-auditory cardiovascular/cerebral adverse health effects by oxidative stress and inflammation. Aircraft noise causes exacerbated adverse effects on blood pressure and endothelial dysfunction in hypertensive mice. Aircraft noise and hypertension potentiate inflammation, ROS formation and oxidative damage in the brain, vessels and heart. Aircraft noise and hypertension seem to have enhanced adverse effects on stress responses in different organs.
Collapse
Affiliation(s)
- Sebastian Steven
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katie Frenis
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sanela Kalinovic
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Miroslava Kvandova
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Johanna Helmstädter
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Omar Hahad
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Konstantina Filippou
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital, Zurich, Switzerland
| | | | - Kerstin Boengler
- Department of Physiology, Justus-Liebig University Gießen, Germany
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Rainer Schulz
- Department of Physiology, Justus-Liebig University Gießen, Germany
| | - Mette Sorensen
- Danish Cancer Society, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Roskilde, Denmark
| | - Andreas Daiber
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
173
|
Batista GMS, Rocha HNM, Storch AS, Garcia VP, Teixeira GF, Mentzinger J, Gomes EAC, Velasco LL, Nóbrega ACL, Rocha NG. Ascorbic acid inhibits vascular remodeling induced by mental stress in overweight/obese men. Life Sci 2020; 250:117554. [PMID: 32184123 DOI: 10.1016/j.lfs.2020.117554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/28/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mental stress (MS) is related to endothelial dysfunction in overweight/obese men. It is believed that the pro-oxidant profile, associated with an imbalance in the vascular remodeling process, may contribute to deleterious effects of MS on endothelial function. However, it is unknown whether administration of ascorbic acid (AA), a potent antioxidant, can prevent oxidative and remodeling dysfunction during MS in these subjects. METHODS Fourteen overweight/obese grade I men (27 ± 7 years; 29.7 ± 2.6 kg·m-2) underwent the Stroop Color Word Test for 5 min to induce MS after AA (3 g) or placebo (PL, 0.9% NaCl) intravenous infusions. Venous blood samples were collected at baseline and the last minute of MS to measure nitrite concentration (chemiluminescence), protein carbonylation, thiobarbituric acid reactive substances (TBARS) and catalase activity (colorimetric assays), superoxide dismutase (SOD; immunoenzymatic assay), activities of active/inactive (pro) forms of metalloproteinases-9 and -2 (MMP; zymography) and its respective tissue inhibitors concentration (TIMP-1 and TIMP-2; immunoenzymatic assays). RESULTS At baseline, MMP-9 activity (p < 0.01), the MMP-9/proMMP-9 ratio (p = 0.02) and TIMP-1 concentration (p = 0.05) were reduced, whereas proMPP-9 activity was increased (p = 0.02) after AA compared to PL infusion. After PL infusion, MS increased protein carbonylation (p < 0.01), catalase (p < 0.01), and the MMP-9/proMMP-9 ratio (p = 0.04) when compared to baseline. AA infusion reduced protein carbonylation (p = 0.02), MMP-9 activity (p < 0.01), and MMP-9/pro-MMP-9 ratio (p < 0.01), while SOD (p = 0.04 vs baseline), proMPP-9 (p < 0.01 vs PL), MMP-2 (p < 0.01 vs PL) and TIMP-2 (p = 0.02 vs baseline) remained elevated during MS. CONCLUSIONS AA appears to minimize the oxidative imbalance and vascular remodeling induced by MS.
Collapse
Affiliation(s)
- G M S Batista
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - H N M Rocha
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - A S Storch
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - V P Garcia
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - G F Teixeira
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - J Mentzinger
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - E A C Gomes
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - L L Velasco
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - A C L Nóbrega
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | - N G Rocha
- Laboratory of Integrative Cardiometabology, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil; Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Brazil.
| |
Collapse
|
174
|
Mouton AJ, Li X, Hall ME, Hall JE. Obesity, Hypertension, and Cardiac Dysfunction: Novel Roles of Immunometabolism in Macrophage Activation and Inflammation. Circ Res 2020; 126:789-806. [PMID: 32163341 PMCID: PMC7255054 DOI: 10.1161/circresaha.119.312321] [Citation(s) in RCA: 352] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and hypertension, which often coexist, are major risk factors for heart failure and are characterized by chronic, low-grade inflammation, which promotes adverse cardiac remodeling. While macrophages play a key role in cardiac remodeling, dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypes promotes excessive inflammation and cardiac injury. Metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation has been implicated in macrophage polarization. M1 macrophages primarily rely on glycolysis, whereas M2 macrophages rely on the tricarboxylic acid cycle and oxidative phosphorylation; thus, factors that affect macrophage metabolism may disrupt M1/M2 homeostasis and exacerbate inflammation. The mechanisms by which obesity and hypertension may synergistically induce macrophage metabolic dysfunction, particularly during cardiac remodeling, are not fully understood. We propose that obesity and hypertension induce M1 macrophage polarization via mechanisms that directly target macrophage metabolism, including changes in circulating glucose and fatty acid substrates, lipotoxicity, and tissue hypoxia. We discuss canonical and novel proinflammatory roles of macrophages during obesity-hypertension-induced cardiac injury, including diastolic dysfunction and impaired calcium handling. Finally, we discuss the current status of potential therapies to target macrophage metabolism during heart failure, including antidiabetic therapies, anti-inflammatory therapies, and novel immunometabolic agents.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Michael E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| |
Collapse
|
175
|
Siedlinski M, Jozefczuk E, Xu X, Teumer A, Evangelou E, Schnabel RB, Welsh P, Maffia P, Erdmann J, Tomaszewski M, Caulfield MJ, Sattar N, Holmes MV, Guzik TJ. White Blood Cells and Blood Pressure: A Mendelian Randomization Study. Circulation 2020; 141:1307-1317. [PMID: 32148083 PMCID: PMC7176352 DOI: 10.1161/circulationaha.119.045102] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND High blood pressure (BP) is a risk factor for cardiovascular morbidity and mortality. While BP is regulated by the function of kidney, vasculature, and sympathetic nervous system, recent experimental data suggest that immune cells may play a role in hypertension. METHODS We studied the relationship between major white blood cell types and blood pressure in the UK Biobank population and used Mendelian randomization (MR) analyses using the ≈750 000 UK-Biobank/International Consortium of Blood Pressure-Genome-Wide Association Studies to examine which leukocyte populations may be causally linked to BP. RESULTS A positive association between quintiles of lymphocyte, monocyte, and neutrophil counts, and increased systolic BP, diastolic BP, and pulse pressure was observed (eg, adjusted systolic BP mean±SE for 1st versus 5th quintile respectively: 140.13±0.08 versus 141.62±0.07 mm Hg for lymphocyte, 139.51±0.08 versus 141.84±0.07 mm Hg for monocyte, and 137.96±0.08 versus 142.71±0.07 mm Hg for neutrophil counts; all P<10-50). Using 121 single nucleotide polymorphisms in MR, implemented through the inverse-variance weighted approach, we identified a potential causal relationship of lymphocyte count with systolic BP and diastolic BP (causal estimates: 0.69 [95% CI, 0.19-1.20] and 0.56 [95% CI, 0.23-0.90] of mm Hg per 1 SD genetically elevated lymphocyte count, respectively), which was directionally concordant to the observational findings. These inverse-variance weighted estimates were consistent with other robust MR methods. The exclusion of rs3184504 SNP in the SH2B3 locus attenuated the magnitude of the signal in some of the MR analyses. MR in the reverse direction found evidence of positive effects of BP indices on counts of monocytes, neutrophils, and eosinophils but not lymphocytes or basophils. Subsequent MR testing of lymphocyte count in the context of genetic correlation with renal function or resting and postexercise heart rate demonstrated a positive association of lymphocyte count with urine albumin-to-creatinine ratio. CONCLUSIONS Observational and genetic analyses demonstrate a concordant, positive and potentially causal relationship of lymphocyte count with systolic BP and diastolic BP.
Collapse
Affiliation(s)
- Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland (M.S., E.J., T.J.G.).,Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| | - Ewelina Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland (M.S., E.J., T.J.G.)
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom (X.X., M.T.)
| | - Alexander Teumer
- Department SHIP/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Germany (A.T.).,German Centre for Cardiovascular Research partner site Greifswald, Germany (A.T.)
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (E.E.)
| | - Renate B Schnabel
- University Heart Center Hamburg Eppendorf, German Center for Cardiovascular Research partner site Hamburg/Kiel/Lübeck, Germany (R.B.S.)
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| | - Pasquale Maffia
- Institute of Infection, Immunity, and Inflammation (P.M.), University of Glasgow, United Kingdom.,Department of Pharmacy, University of Naples Federico II, Italy (P.M.)
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Germany (J.E.)
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom (X.X., M.T.)
| | - Mark J Caulfield
- William Harvey Research Institute, National Institute for Health Research Biomedical Research Centre at Barts, Queen Mary University of London, United Kingdom (M.J.C.)
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom (M.V.H.)
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland (M.S., E.J., T.J.G.).,Institute of Cardiovascular and Medical Sciences (M.S., P.W., N.S., T.J.G.), University of Glasgow, United Kingdom
| |
Collapse
|
176
|
Drummond GR, Vinh A, Guzik TJ, Sobey CG. Immune mechanisms of hypertension. Nat Rev Immunol 2020; 19:517-532. [PMID: 30992524 DOI: 10.1038/s41577-019-0160-5] [Citation(s) in RCA: 265] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension affects 30% of adults and is the leading risk factor for heart attack and stroke. Traditionally, hypertension has been regarded as a disorder of two systems that are involved in the regulation of salt-water balance and cardiovascular function: the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system (SNS). However, current treatments that aim to limit the influence of the RAAS or SNS on blood pressure fail in ~40% of cases, which suggests that other mechanisms must be involved. This Review summarizes the clinical and experimental evidence supporting a contribution of immune mechanisms to the development of hypertension. In this context, we highlight the immune cell subsets that are postulated to either promote or protect against hypertension through modulation of cardiac output and/or peripheral vascular resistance. We conclude with an appraisal of knowledge gaps still to be addressed before immunomodulatory therapies might be applied to at least a subset of patients with hypertension.
Collapse
Affiliation(s)
- Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia.
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Tomasz J Guzik
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland.,BHF Centre of Research Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
177
|
Lang PP, Bai J, Zhang YL, Yang XL, Xia YL, Lin QY, Li HH. Blockade of intercellular adhesion molecule-1 prevents angiotensin II-induced hypertension and vascular dysfunction. J Transl Med 2020; 100:378-386. [PMID: 31527830 DOI: 10.1038/s41374-019-0320-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 12/31/2022] Open
Abstract
Monocyte and adhesion infiltration into the arterial subendothelium are initial steps in hypertension development. The endothelial intercellular adhesion molecule-1 (ICAM-1) has been implicated in the recruitment and adhesion of leukocytes in several cardiac diseases. However, the role of ICAM-1 in angiotensin II (Ang II)-induced hypertension development remains unknown. Hypertension was induced by administering an infusion of Ang II (1000 ng/kg/min) to wild-type (WT) mice treated with an IgG control or ICAM-1 neutralizing antibody (1 and 2 mg/mouse/day, respectively). Blood pressure was determined using the tail-cuff system. Vascular remodeling was assessed by performing a histological examination. Inflammation and reactive oxygen species (ROS) levels were determined by using immunostaining. Vascular dysfunction was assessed by aortic ring assay. The expression of fibrotic markers, cytokines and NOX was evaluated by quantitative real-time PCR analysis. Our results demonstrate that Ang II infusion markedly increased the ICAM-1 level in the aorta. Blocking ICAM-1 with a neutralizing antibody significantly attenuated Ang II-induced arterial hypertension, vascular hypertrophy, fibrosis, macrophage infiltration, and ROS production and improved vascular relaxation. In conclusion, ICAM-1-mediated monocyte adhesion and migration play a critical role in Ang II-induced arterial hypertension and vascular dysfunction. ICAM-1 inhibitors may represent a new therapeutic strategy for the treatment of this disease.
Collapse
Affiliation(s)
- Ping-Ping Lang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Jie Bai
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Yun-Long Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Xiao-Lei Yang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Yun-Long Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Qiu-Yue Lin
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China.
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China.
| |
Collapse
|
178
|
Carnevale D, Wenzel P. Mechanical stretch on endothelial cells interconnects innate and adaptive immune response in hypertension. Cardiovasc Res 2020; 114:1432-1434. [PMID: 29912294 DOI: 10.1093/cvr/cvy148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, Pozzilli, IS, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Philip Wenzel
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstrasse 1, Mainz, Germany.,Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, Germany
| |
Collapse
|
179
|
Masenga SK, Elijovich F, Hamooya BM, Nzala S, Kwenda G, Heimburger DC, Mutale W, Munsaka SM, Zhao S, Koethe JR, Kirabo A. Elevated Eosinophils as a Feature of Inflammation Associated With Hypertension in Virally Suppressed People Living With HIV. J Am Heart Assoc 2020; 9:e011450. [PMID: 32064996 PMCID: PMC7070208 DOI: 10.1161/jaha.118.011450] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background People living with HIV (PLWH) are at increased risk of cardiovascular disease, including hypertension, which persists despite effective plasma viral suppression on antiretroviral therapy. HIV infection is characterized by long‐term alterations in immune function, but the contribution of immune factors to hypertension in PLWH is not fully understood. Prior studies have found that both innate and adaptive immune cell activation contributes to hypertension. Methods and Results We hypothesized that chronic inflammation may contribute to hypertension in PLWH. To test this hypothesis, we enrolled a cohort of 70 PLWH (44% hypertensive) on a long‐term single antiretroviral therapy regimen for broad phenotyping of inflammation biomarkers. We found that hypertensive PLWH had higher levels of inflammatory cytokines, including tumor necrosis factor‐α receptor 1, interleukin‐6, interleukin‐17, interleukin‐5, intercellular adhesion molecule 1 and macrophage inflammatory protein‐1α. After adjustment for age, sex, and fat mass index, the circulating eosinophils remained significantly associated with hypertension. On the basis of these results, we assessed the relationship of eosinophils and hypertension in 2 cohorts of 50 and 81 039 similar HIV‐negative people; although eosinophil count was associated with prevalent hypertension, this relationship was abrogated by body mass index. Conclusions These findings may represent a unique linkage between immune status and cardiovascular physiological characteristics in HIV infection, which should be evaluated further.
Collapse
Affiliation(s)
- Sepiso K Masenga
- School of Medicine and Health Sciences Mulungushi University Livingstone Zambia.,Department of Biomedical Sciences School of Health Sciences University of Zambia Lusaka Zambia.,Vanderbilt Institute for Global Health Vanderbilt University Medical Center Nashville TN
| | - Fernando Elijovich
- Division of Clinical Pharmacology Vanderbilt University Medical Center Nashville TN
| | - Benson M Hamooya
- School of Medicine and Health Sciences Mulungushi University Livingstone Zambia.,Department of Epidemiology and Biostatistics School of Public Health University of Zambia Lusaka Zambia
| | - Selestine Nzala
- Department of Medical Education Development University of Zambia Lusaka Zambia
| | - Geoffrey Kwenda
- Department of Biomedical Sciences School of Health Sciences University of Zambia Lusaka Zambia
| | - Douglas C Heimburger
- Vanderbilt Institute for Global Health Vanderbilt University Medical Center Nashville TN
| | - Wilbroad Mutale
- Department of Health Policy and Management School of Public Health University of Zambia Lusaka Zambia
| | - Sody M Munsaka
- Department of Biomedical Sciences School of Health Sciences University of Zambia Lusaka Zambia
| | - Shilin Zhao
- Department of Biostatistics Vanderbilt University Medical Center Nashville TN
| | - John R Koethe
- Division of Infectious Diseases Vanderbilt University Medical Center Nashville TN
| | - Annet Kirabo
- Division of Clinical Pharmacology Vanderbilt University Medical Center Nashville TN.,Department of Molecular Physiology and Biophysics Vanderbilt University Nashville TN
| |
Collapse
|
180
|
Nosalski R, Siedlinski M, Denby L, McGinnigle E, Nowak M, Cat AND, Medina-Ruiz L, Cantini M, Skiba D, Wilk G, Osmenda G, Rodor J, Salmeron-Sanchez M, Graham G, Maffia P, Graham D, Baker AH, Guzik TJ. T-Cell-Derived miRNA-214 Mediates Perivascular Fibrosis in Hypertension. Circ Res 2020; 126:988-1003. [PMID: 32065054 PMCID: PMC7147427 DOI: 10.1161/circresaha.119.315428] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [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
RATIONALE Despite increasing understanding of the prognostic importance of vascular stiffening linked to perivascular fibrosis in hypertension, the molecular and cellular regulation of this process is poorly understood. OBJECTIVES To study the functional role of microRNA-214 (miR-214) in the induction of perivascular fibrosis and endothelial dysfunction driving vascular stiffening. METHODS AND RESULTS Out of 381 miRs screened in the perivascular tissues in response to Ang II (angiotensin II)-mediated hypertension, miR-214 showed the highest induction (8-fold, P=0.0001). MiR-214 induction was pronounced in perivascular and circulating T cells, but not in perivascular adipose tissue adipocytes. Global deletion of miR-214-/- prevented Ang II-induced periaortic fibrosis, Col1a1, Col3a1, Col5a1, and Tgfb1 expression, hydroxyproline accumulation, and vascular stiffening, without difference in blood pressure. Mechanistic studies revealed that miR-214-/- mice were protected against endothelial dysfunction, oxidative stress, and increased Nox2, all of which were induced by Ang II in WT mice. Ang II-induced recruitment of T cells into perivascular adipose tissue was abolished in miR-214-/- mice. Adoptive transfer of miR-214-/- T cells into RAG1-/- mice resulted in reduced perivascular fibrosis compared with the effect of WT T cells. Ang II induced hypertension caused significant change in the expression of 1380 T cell genes in WT, but only 51 in miR-214-/-. T cell activation, proliferation and chemotaxis pathways were differentially affected. MiR-214-/- prevented Ang II-induction of profibrotic T cell cytokines (IL-17, TNF-α, IL-9, and IFN-γ) and chemokine receptors (CCR1, CCR2, CCR4, CCR5, CCR6, and CXCR3). This manifested in reduced in vitro and in vivo T cell chemotaxis resulting in attenuation of profibrotic perivascular inflammation. Translationally, we show that miR-214 is increased in plasma of patients with hypertension and is directly correlated to pulse wave velocity as a measure of vascular stiffness. CONCLUSIONS T-cell-derived miR-214 controls pathological perivascular fibrosis in hypertension mediated by T cell recruitment and local profibrotic cytokine release.
Collapse
Affiliation(s)
- Ryszard Nosalski
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.).,Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| | - Mateusz Siedlinski
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| | - Laura Denby
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, United Kingdom (L.D., J.R., A.H.B.)
| | - Eilidh McGinnigle
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.)
| | - Michal Nowak
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| | - Aurelie Nguyen Dinh Cat
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.)
| | - Laura Medina-Ruiz
- Institute of Infection, Immunity and Inflammation, University of Glasgow, United Kingdom (L.M.-R., G.G., P.M.)
| | - Marco Cantini
- Centre for the Cellular Microenvironment, School of Engineering, University of Glasgow, United Kingdom (M.C., M.S.-S.)
| | - Dominik Skiba
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.).,Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| | - Grzegorz Wilk
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| | - Grzegorz Osmenda
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| | - Julie Rodor
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, United Kingdom (L.D., J.R., A.H.B.)
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, School of Engineering, University of Glasgow, United Kingdom (M.C., M.S.-S.)
| | - Gerard Graham
- Institute of Infection, Immunity and Inflammation, University of Glasgow, United Kingdom (L.M.-R., G.G., P.M.)
| | - Pasquale Maffia
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.).,Institute of Infection, Immunity and Inflammation, University of Glasgow, United Kingdom (L.M.-R., G.G., P.M.).,Department of Pharmacy, University of Naples Federico II, Italy (P.M.)
| | - Delyth Graham
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.)
| | - Andrew H Baker
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, United Kingdom (L.D., J.R., A.H.B.)
| | - Tomasz J Guzik
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.N., E.M., A.N.D.C., D.S., P.M., D.G., T.J.G.).,Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (R.N., M.S., M.N., D.S., G.W., G.O., T.J.G.)
| |
Collapse
|
181
|
Zhu X, Wang Y, Zhu L, Zhu Y, Zhang K, Wang L, Bai H, Yang Q, Ben J, Zhang H, Li X, Xu Y, Chen Q. Class A1 scavenger receptor prevents obesity-associated blood pressure elevation through suppressing overproduction of vascular endothelial growth factor B in macrophages. Cardiovasc Res 2020; 117:547-560. [PMID: 32044963 DOI: 10.1093/cvr/cvaa030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/17/2019] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
AIMS Dysfunctional innate immune function and inflammation contributes to the pathogenesis of obesity-associated hypertension, in which macrophage infiltration in the perivascular adipose tissue (PVAT) plays a key role. However, the mechanisms behind it are not well understood. Class A1 scavenger receptor (SR-A1) is one of the major pattern recognition receptors in modulating macrophage activity, and here, we aimed to investigate its role in obesity-associated hypertension. METHODS AND RESULTS Both diet-induced and genetic obesity were generated in mice. Deficiency in SR-A1 aggravated the obesity-induced blood pressure (BP) elevation and endothelial dysfunction in mice. The BP-elevating effect of SR-A1 deficiency was blocked by the down-regulation of vascular endothelial growth factor B (VEGF-B) in obese mice. Overexpression of VEGF-B raised BP in the obese mice but not in normal mice. Administration of fucoidan, a ligand of SR-A1, lowered BP, and VEGF-B levels in Sr-a1+/+ but not in Sr-a1-/- obese mice. CONCLUSION These results reveal a new link between PVAT and vascular biology in obesity orchestrated by the SR-A1/VEGF-B axis in macrophages. SR-A1 and VEGF-B may be promising therapeutic targets in the treatment of obesity-associated hypertension.
Collapse
Affiliation(s)
- Xudong Zhu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Yan Wang
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Liu Zhu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China.,Cardiovascular Medicine Department, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Gusu District, Suzhou 215004, China
| | - Ye Zhu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Kun Zhang
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Lei Wang
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Hui Bai
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Qing Yang
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Jingjing Ben
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Hanwen Zhang
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Xiaoyu Li
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Yong Xu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| | - Qi Chen
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Liongmian Road, Jiangning District, Nanjing 211166, China
| |
Collapse
|
182
|
Schiffrin EL. How Structure, Mechanics, and Function of the Vasculature Contribute to Blood Pressure Elevation in Hypertension. Can J Cardiol 2020; 36:648-658. [PMID: 32389338 DOI: 10.1016/j.cjca.2020.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 01/11/2023] Open
Abstract
Large conduit arteries and the microcirculation participate in the mechanisms of elevation of blood pressure (BP). Large vessels play roles predominantly in older subjects, with stiffening progressing after middle age leading to increases in systolic BP found in most humans with aging. Systolic BP elevation and increased pulsatility penetrate deeper into the distal vasculature, leading to microcirculatory injury, remodelling, and associated endothelial dysfunction. The result is target organ damage in the heart, brain, and kidney. In younger individuals genetically predisposed to high BP, increased salt intake or other exogenous or endogenous risk factors for hypertension, including overweight and excess alcohol intake, lead to enhanced sympathetic activity and vasoconstriction. Enhanced vasoconstrictor responses and myogenic tone become persistent when embedded in an increased extracellular matrix, resulting in remodelling of resistance arteries with a narrowed lumen and increased media-lumen ratio. Stimulation of the renin-angiotensin-aldosterone and endothelin systems and inflammatory and immune activation, to which gut microbiome dysbiosis may contribute as a result of salt intake, also participate in the injury and remodelling of the microcirculation and endothelial dysfunction. Inflammation of perivascular fat and loss of anticontractile factors play roles as well in microvessel remodelling. Exaggerated myogenic tone leads to closure of terminal arterioles, collapse of capillaries and venules, functional rarefaction, and eventually to anatomic rarefaction, compromising tissue perfusion. The remodelling of the microcirculation raises resistance to flow, and accordingly raises BP in a feedback process that over years results in stiffening of conduit arteries and systo-diastolic or predominantly systolic hypertension and, more rarely, predominantly diastolic hypertension. Thus, at different stages of life and the evolution of hypertension, large vessels and the microcirculation interact to contribute to BP elevation.
Collapse
Affiliation(s)
- Ernesto L Schiffrin
- Lady Davis Institute for Medical Research and Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
183
|
Xu J, Sun Y, Lu J. Knockdown of Long Noncoding RNA (lncRNA) AK094457 Relieved Angiotensin II Induced Vascular Endothelial Cell Injury. Med Sci Monit 2020; 26:e919854. [PMID: 32027625 PMCID: PMC7020760 DOI: 10.12659/msm.919854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Hypertension could induce many serious diseases, including damage to vascular endothelial cells. As a non-coding RNA, long noncoding RNA (lncRNA) has received much attention in scientific research and has a regulating efficacy on many critical life activities in human body. The level of lncRNA AK094457 is thought to be elevated in hypertensive rats. However, there is no research indicating the relationship between the level of lncRNA AK094457 and vascular endothelial injury. Material/Methods In our study, we used lentiviral to knockdown lncRNA AK094457, and the human umbilical vein endothelial cells (HUVECs) were stimulated by the Ang II to imitate the vascular endothelial cell damage caused by hypertension. The Cell Counting Kit-8 assays were used to detect the cells viability. Western blotting was performed to detect the endothelial nitric oxide synthase (eNOS), p-eNOS and endothelin-1 (ET-1). After that the production of the NO was monitored. At last, the reactive oxygen species (ROS) levels and apoptosis rates were detected in this study. Results According to the results, we found that knockdown lncRNA AK094457 could alleviate the decrease of vascular endothelial cell viability induced by angiotensin II (Ang II). The knockdown of lncRNA AK094457 also relieved the downregulation of eNOS and p-eNOS, and the decreasing of NO release. At the same time, the knockdown of lncRNA inhibited the levels of Ang II-induced proinflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1, and IL-6) and cell adhesion molecules (vascular cell adhesion molecule 1 [VCAM-1], intercellular adhesion molecule 1 [ICAM-1], and monocyte chemoattractant protein-1 [MCP-1]). The levels of ROS and apoptosis rates also decreased after the knockdown of lncRNA AK094457. Conclusions All these results indicated that lncRNA AK094457 could promote Ang II-induced vascular endothelial cell injury. On the contrary, knockdown of lncRNA AK094457 could alleviate this damage.
Collapse
Affiliation(s)
- JiaYi Xu
- Department of Gerontology, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| | - Yingjie Sun
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| | - Jie Lu
- Department of Gerontology, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
184
|
Justina VD, Giachini FR, Priviero F, Webb RC. Double-stranded RNA and Toll-like receptor activation: a novel mechanism for blood pressure regulation. Clin Sci (Lond) 2020; 134:303-313. [PMID: 31998948 PMCID: PMC7703673 DOI: 10.1042/cs20190913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/24/2022]
Abstract
Toll-like receptors (TLRs), such as TLR4 and 9, recognize pathogen-associated molecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) and are associated with increased blood pressure (BP). TLR3, residing in the endosomal compartment, is activated by viral double-stranded RNA (dsRNA) leading to activation of TIR receptor domain-containing adaptor inducing IFN-β (TRIF) dependent pathway. Besides foreign pathogens, the immune system responds to endogenous markers of cellular damage such as mitochondrial dsRNA (mtdsRNA). New evidence has shown a link between dsRNA and increased BP. Moreover, TLR3 activation during pregnancy was demonstrated to develop preeclampsia-like symptoms in both rats and mice. Hence, we hypothesize that the dsRNA derived from viral nucleic acids or cellular damage (mtdsRNA) will increase the inflammatory state through activation of TLR3, contributing to vascular dysfunction and increased BP. Therefore, inhibition of TLR3 could be a therapeutic target for the treatment of hypertension with potential improvement in vascular reactivity and consequently, a decrease in BP.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Brazil
| | - Fernanda R. Giachini
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Brazil
- RIVATREM - Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo
| | - Fernanda Priviero
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - R. Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| |
Collapse
|
185
|
Xiao L, Harrison DG. Inflammation in Hypertension. Can J Cardiol 2020; 36:635-647. [PMID: 32389337 DOI: 10.1016/j.cjca.2020.01.013] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/26/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022] Open
Abstract
For more than 50 years, evidence has accumulated that inflammation contributes to the pathogenesis of hypertension. Immune cells have been observed in vessels and kidneys of hypertensive humans. Biomarkers of inflammation, including high sensitivity C-reactive protein, various cytokines, and products of the complement pathway are elevated in humans with hypertension. Emerging evidence suggests that hypertension is accompanied and indeed initiated by activation of complement, the inflammasome, and by a change in the phenotype of circulating immune cells, particularly myeloid cells. High-dimensional transcriptomic analyses are providing insight into new subclasses of immune cells that are likely injurious in hypertension. These inflammatory events are interdependent and there is ultimately engagement of the adaptive immune system through mechanisms involving oxidative stress, modification of endogenous proteins, and alterations in antigen processing and presentation. These observations suggest new therapeutic opportunities to reduce end organ damage in hypertension might be used and guided by levels of inflammatory biomarkers.
Collapse
Affiliation(s)
- Liang Xiao
- Departments of Medicine, Pharmacology, and Physiology, and Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - David G Harrison
- Departments of Medicine, Pharmacology, and Physiology, and Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
186
|
|
187
|
Kura B, Szeiffova Bacova B, Kalocayova B, Sykora M, Slezak J. Oxidative Stress-Responsive MicroRNAs in Heart Injury. Int J Mol Sci 2020; 21:E358. [PMID: 31948131 PMCID: PMC6981696 DOI: 10.3390/ijms21010358] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are important molecules in the living organisms as a part of many signaling pathways. However, if overproduced, they also play a significant role in the development of cardiovascular diseases, such as arrhythmia, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction and heart transplantation), and heart failure. As a result of oxidative stress action, apoptosis, hypertrophy, and fibrosis may occur. MicroRNAs (miRNAs) represent important endogenous nucleotides that regulate many biological processes, including those involved in heart damage caused by oxidative stress. Oxidative stress can alter the expression level of many miRNAs. These changes in miRNA expression occur mainly via modulation of nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, calcineurin/nuclear factor of activated T cell (NFAT), or nuclear factor kappa B (NF-κB) pathways. Up until now, several circulating miRNAs have been reported to be potential biomarkers of ROS-related cardiac diseases, including myocardial infarction, hypertrophy, ischemia/reperfusion, and heart failure, such as miRNA-499, miRNA-199, miRNA-21, miRNA-144, miRNA-208a, miRNA-34a, etc. On the other hand, a lot of studies are aimed at using miRNAs for therapeutic purposes. This review points to the need for studying the role of redox-sensitive miRNAs, to identify more effective biomarkers and develop better therapeutic targets for oxidative-stress-related heart diseases.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| |
Collapse
|
188
|
Zhang N, Zhang Y, Wu B, You S, Sun Y. Role of WW domain E3 ubiquitin protein ligase 2 in modulating ubiquitination and Degradation of Septin4 in oxidative stress endothelial injury. Redox Biol 2020; 30:101419. [PMID: 31924572 PMCID: PMC6951091 DOI: 10.1016/j.redox.2019.101419] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/18/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress-associated endothelial injury is the initial event and major cause of multiple cardiovascular diseases such as atherosclerosis and hypertensive angiopathy. A protein homeostasis imbalance is a critical cause of endothelial injury, and homologous to E6AP C-terminus (HECT)-type E3 ubiquitin ligases are the core factors controlling protein homeostasis. Although HECT-type E3 ubiquitin ligases are involved in the regulation of cardiac development and diseases, their roles in endothelial injury remain largely unknown. This study aimed to identify which HECT-type E3 ubiquitin ligase is involved in endothelial injury and clarify the mechanisms at molecular, cellular, and organism levels. We revealed a novel role of the HECT-type E3 ubiquitin ligase WWP2 in regulating endothelial injury and vascular remodeling after endothelial injury. Endothelial/myeloid-specific WWP2 knockout in mice significantly aggravated angiotensin II/oxidative stress-induced endothelial injury and vascular remodeling after endothelial injury. The same results were obtained from in vitro experiments. Mechanistically, the endothelial injury factor Septin4 was identified as a novel physiological substrate of WWP2. In addition, WWP2 interacted with the GTPase domain of Septin4, ubiquitinating Septin4-K174 to degrade Septin4 through the ubiquitin-proteasome system, which inhibited the Septin4-PARP1 endothelial damage complex. These results identified the first endothelial injury-associated physiological pathway regulated by HECT-type E3 ubiquitin ligases in vivo as well as a unique proteolytic mechanism through which WWP2 controls endothelial injury and vascular remodeling after endothelial injury. These findings might provide a novel treatment strategy for oxidative stress-associated atherosclerosis and hypertensive vascular diseases.
Collapse
Affiliation(s)
- Naijin Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ying Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Boquan Wu
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Shilong You
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
189
|
An X, Jiang G, Cheng C, Lv Z, Liu Y, Wang F. Inhibition of Platelets by Clopidogrel Suppressed Ang II-Induced Vascular Inflammation, Oxidative Stress, and Remodeling. J Am Heart Assoc 2019; 7:e009600. [PMID: 30608200 PMCID: PMC6404205 DOI: 10.1161/jaha.118.009600] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Platelets play a role in promoting inflammatory responses under several disease conditions. Platelets are activated in hypertensive patients. However, the mechanisms responsible for platelet‐mediating vascular inflammation are unknown. The present study investigated the role of platelets in promoting vascular inflammation following angiotensin II (Ang II) stimulation, and the efficacy of antiplatelet intervention. Methods and Results Within a mouse model of Ang II infusion (490 ng/kg per min), we measured the portion of P‐selectin–positive platelets and platelet‐monocyte (P‐M) binding in blood samples, and platelet accumulation and P‐M binding in vessels under Ang II stimulation at days 1, 3, and 7. We tested the efficacy of clopidogrel (15 mg/kg per day, followed by 5 mg/kg per day) on Ang II‐induced platelet activation, P‐M binding, vascular platelet accumulation, as well as vascular inflammation and remodeling at day 7 or 14. Clopidogrel reduced platelet vascular deposition (28.7±2.4% versus 18.3±2.9%), suppressed inflammatory cell infiltration (3.6±0.8×104/vessel versus 2.3±1.2×104/vessel) and oxidative stress, and attenuated vascular remodeling and dysfunction (55.0±5.5% versus 84.0±6.0%) following Ang II stimulation at day 7 or 14. Clopidogrel suppressed Ang II‐induced P‐M binding both at circulating (13.4±3.3% versus 5.9±2.7%) and regional (33.4±4.3% versus 11.9±2.7%) levels. Conclusions Platelets play a critical role in vascular inflammation under Ang II stimulation, with a marked promotion of P‐M binding as an important mechanism. Clopidogrel prevented vascular inflammation in Ang II‐infused mice.
Collapse
Affiliation(s)
- Xiangbo An
- 1 Department of Interventional Therapy the First Affiliated Hospital of Dalian Medical University Dalian China.,3 Institute of Cardiovascular Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Guinan Jiang
- 1 Department of Interventional Therapy the First Affiliated Hospital of Dalian Medical University Dalian China.,3 Institute of Cardiovascular Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Cheng Cheng
- 4 Center for Clinical Research on Neurological Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Zhengshuai Lv
- 2 Department of Anesthesia the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Yang Liu
- 3 Institute of Cardiovascular Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Feng Wang
- 1 Department of Interventional Therapy the First Affiliated Hospital of Dalian Medical University Dalian China
| |
Collapse
|
190
|
Helmstädter J, Frenis K, Filippou K, Grill A, Dib M, Kalinovic S, Pawelke F, Kus K, Kröller-Schön S, Oelze M, Chlopicki S, Schuppan D, Wenzel P, Ruf W, Drucker DJ, Münzel T, Daiber A, Steven S. Endothelial GLP-1 (Glucagon-Like Peptide-1) Receptor Mediates Cardiovascular Protection by Liraglutide In Mice With Experimental Arterial Hypertension. Arterioscler Thromb Vasc Biol 2019; 40:145-158. [PMID: 31747801 PMCID: PMC6946108 DOI: 10.1161/atv.0000615456.97862.30] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Supplemental Digital Content is available in the text. Cardiovascular outcome trials demonstrated that GLP-1 (glucagon-like peptide-1) analogs including liraglutide reduce the risk of cardiovascular events in type 2 diabetes mellitus. Whether GLP-1 analogs reduce the risk for atherosclerosis independent of glycemic control is challenging to elucidate as the GLP-1R (GLP-1 receptor) is expressed on different cell types, including endothelial and immune cells.
Collapse
Affiliation(s)
- Johanna Helmstädter
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katie Frenis
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Konstantina Filippou
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Alexandra Grill
- Center for Thrombosis and Hemostasis (A.G., P.W., W.R., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (A.G., W.R., T.M., A.D.)
| | - Mobin Dib
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sanela Kalinovic
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Franziska Pawelke
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET) (K.K., S.C.), Jagiellonian University, Krakow, Poland
| | - Swenja Kröller-Schön
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Oelze
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET) (K.K., S.C.), Jagiellonian University, Krakow, Poland.,Chair of Pharmacology (S.C.), Jagiellonian University, Krakow, Poland
| | - Detlef Schuppan
- Institute of Translational Immunology (D.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Philip Wenzel
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Center for Thrombosis and Hemostasis (A.G., P.W., W.R., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (A.G., P.W., W.R., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (A.G., W.R., T.M., A.D.)
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Canada (D.J.D.)
| | - Thomas Münzel
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (A.G., W.R., T.M., A.D.)
| | - Andreas Daiber
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (A.G., W.R., T.M., A.D.)
| | - Sebastian Steven
- From the Center for Cardiology (J.H., K. Frenis, K. Filippou, M.D., S.K., F.P., S.K.-S., M.O., P.W. T.M., A.D., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Center for Thrombosis and Hemostasis (A.G., P.W., W.R., S.S.), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
191
|
Xiao L, Itani HA, do Carmo LS, Carver LS, Breyer RM, Harrison DG. Central EP3 (E Prostanoid 3) Receptors Mediate Salt-Sensitive Hypertension and Immune Activation. Hypertension 2019; 74:1507-1515. [PMID: 31679420 PMCID: PMC7040566 DOI: 10.1161/hypertensionaha.119.13850] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We recently identified a pathway underlying immune activation in hypertension. Proteins oxidatively modified by reactive isoLG (isolevuglandin) accumulate in dendritic cells (DCs). PGE2 (Prostaglandin E2) has been implicated in the inflammation associated with hypertension. We hypothesized that PGE2 via its EP (E prostanoid) 3 receptor contributes to DC activation in hypertension. EP3-/- mice and wild-type littermates were exposed to sequential hypertensive stimuli involving an initial 2-week exposure to the nitric oxide synthase inhibitor Nω-nitro-L-arginine methyl ester hydrochloride in drinking water, followed by a 2-week washout period, and a subsequent 4% high-salt diet for 3 weeks. In wild-type mice, this protocol increased systolic pressure from 123±2 to 148±8 mm Hg (P<0.05). This was associated with marked renal inflammation and a striking accumulation of isoLG adducts in splenic DCs. However, the increases in blood pressure, renal T-cell infiltration, and DC isoLG formation were completely prevented in EP3-/- mice. Similar protective effects were also observed in wild-type mice that received intracerebroventricular injection of a lentiviral vector encoding shRNA targeting the EP3 receptor. Further, in vitro experiments indicated that PGE2 also acts directly on DCs via its EP1 receptors to stimulate intracellular isoLG formation. Together, these findings provide new insight into how EP receptors in both the central nervous system and peripherally on DCs promote inflammation in salt-induced hypertension.
Collapse
Affiliation(s)
- Liang Xiao
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Hana A Itani
- Department of Pharmacology and Toxicology, American University of Beirut, Lebanon (H.A.I.)
| | - Luciana Simao do Carmo
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lucas S Carver
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Richard M Breyer
- Division of Nephrology and Hypertension (R.M.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - David G Harrison
- From the Division of Clinical Pharmacology (L.X., L.S.d.C., L.C., D.G.H.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
192
|
Wenstedt EF, Verberk SG, Kroon J, Neele AE, Baardman J, Claessen N, Pasaoglu ÖT, Rademaker E, Schrooten EM, Wouda RD, de Winther MP, Aten J, Vogt L, Van den Bossche J. Salt increases monocyte CCR2 expression and inflammatory responses in humans. JCI Insight 2019; 4:130508. [PMID: 31672939 DOI: 10.1172/jci.insight.130508] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/26/2019] [Indexed: 01/03/2023] Open
Abstract
Inflammation may play a role in the link between high salt intake and its deleterious consequences. However, it is unknown whether salt can induce proinflammatory priming of monocytes and macrophages in humans. We investigated the effects of salt on monocytes and macrophages in vitro and in vivo by performing a randomized crossover trial in which 11 healthy human subjects adhered to a 2-week low-salt and high-salt diet. We demonstrate that salt increases monocyte expression of CCR2, a chemokine receptor that mediates monocyte infiltration in inflammatory diseases. In line with this, we show a salt-induced increase of plasma MCP-1, transendothelial migration of monocytes, and skin macrophage density after high-salt diet. Macrophages demonstrate signs of an increased proinflammatory phenotype after salt exposure, as represented by boosted LPS-induced cytokine secretion of IL-6, TNF, and IL-10 in vitro, and by increased HLA-DR expression and decreased CD206 expression on skin macrophages after high-salt diet. Taken together, our data open up the possibility for inflammatory monocyte and macrophage responses as potential contributors to the deleterious effects of high salt intake.
Collapse
Affiliation(s)
- Eliane Fe Wenstedt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Sanne Gs Verberk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Amsterdam UMC, University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Annette E Neele
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Section of Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Jeroen Baardman
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Section of Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Nike Claessen
- Amsterdam UMC, University of Amsterdam, Department of Pathology, Amsterdam, Netherlands
| | - Özge T Pasaoglu
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Emma Rademaker
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Esmee M Schrooten
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Rosa D Wouda
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Menno Pj de Winther
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Section of Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention (IPEK), Munich, Germany
| | - Jan Aten
- Amsterdam UMC, University of Amsterdam, Department of Pathology, Amsterdam, Netherlands
| | - Liffert Vogt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Section of Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| |
Collapse
|
193
|
Jansen T, Kröller-Schön S, Schönfelder T, Foretz M, Viollet B, Daiber A, Oelze M, Brandt M, Steven S, Kvandová M, Kalinovic S, Lagrange J, Keaney JF, Münzel T, Wenzel P, Schulz E. α1AMPK deletion in myelomonocytic cells induces a pro-inflammatory phenotype and enhances angiotensin II-induced vascular dysfunction. Cardiovasc Res 2019; 114:1883-1893. [PMID: 29982418 DOI: 10.1093/cvr/cvy172] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/02/2018] [Indexed: 12/17/2022] Open
Abstract
Aims Immune cell function involves energy-dependent processes including growth, proliferation, and cytokine production. Since the AMP-activated protein kinase (AMPK) is a crucial regulator of intracellular energy homeostasis, its expression and activity may also affect innate and adaptive immune cell responses. Therefore, we aimed to investigate the consequences of α1AMPK deletion in myelomonocytic cells on vascular function, inflammation, and hypertension during chronic angiotensin II (ATII) treatment. Methods and results We generated a mouse strain with α1AMPK deletion in lysozyme M+ myelomonocytic cells. Compared to controls, chronic ATII infusion (1 mg/kg/day for 7 days) lead to increased vascular oxidative stress and aggravated endothelial dysfunction in LysM-Cre+ x α1AMPKfl/fl mice. This was accompanied by an increased aortic infiltration of CD11b+F4/80+ macrophages and enhanced pro-inflammatory cytokine release (tumour necrosis factor-alpha, interferon-gamma, and interleukin-6). Mechanistically, we found that increased expression of C-C chemokine receptor 2 (CCR2) in α1AMPK deficient myelomonocytic cells facilitated their recruitment to the vascular wall. In addition, expression of the ATII receptor type 1a and the oxidative burst was increased in these cells, indicating an increased susceptibility towards pro-oxidant stimuli. Conclusions In summary, α1AMPK deletion in myelomonocytic cells aggravates vascular oxidative stress and dysfunction by enhancing their recruitment to the vascular wall and increasing their susceptibility towards pro-oxidant stimuli. Our observations suggest that metabolic control in myelomonocytic cells has profound implications for their inflammatory phenotype and may trigger the development of vascular disease.
Collapse
Affiliation(s)
- Thomas Jansen
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Swenja Kröller-Schön
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Tanja Schönfelder
- Center for Thrombosis and Hemostasis (CTH), Universitätsmedizin Mainz, Mainz, Germany
| | - Marc Foretz
- Institut Cochin, INSERM U1016, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris cité, 24 rue du faubourg Saint Jacques, Paris, France
| | - Benoit Viollet
- Institut Cochin, INSERM U1016, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris cité, 24 rue du faubourg Saint Jacques, Paris, France
| | - Andreas Daiber
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Matthias Oelze
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Moritz Brandt
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Universitätsmedizin Mainz, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Universitätsmedizin Mainz, Mainz, Germany
| | - Miroslava Kvandová
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Sanela Kalinovic
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Jeremy Lagrange
- Center for Thrombosis and Hemostasis (CTH), Universitätsmedizin Mainz, Mainz, Germany
| | - John F Keaney
- Division of Cardiovascular Medicine, UMass Medical School, 55 Lake Avenue North, Worcester, MA, USA
| | - Thomas Münzel
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - Philip Wenzel
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Universitätsmedizin Mainz, Mainz, Germany
| | - Eberhard Schulz
- Department of Cardiology 1, Center for Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| |
Collapse
|
194
|
Fang P, Li X, Shan H, Saredy JJ, Cueto R, Xia J, Jiang X, Yang XF, Wang H. Ly6C + Inflammatory Monocyte Differentiation Partially Mediates Hyperhomocysteinemia-Induced Vascular Dysfunction in Type 2 Diabetic db/db Mice. Arterioscler Thromb Vasc Biol 2019; 39:2097-2119. [PMID: 31366217 PMCID: PMC6761027 DOI: 10.1161/atvbaha.119.313138] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Hyperhomocysteinemia (HHcy) is a potent risk factor for diabetic cardiovascular diseases. We have previously reported that hyperhomocysteinemia potentiates type 1 diabetes mellitus-induced inflammatory monocyte differentiation, vascular dysfunction, and atherosclerosis. However, the effects of hyperhomocysteinemia on vascular inflammation in type 2 diabetes mellitus (T2DM) and the underlying mechanism are unknown. Approach and Results: Here, we demonstrate that hyperhomocysteinemia was induced by a high methionine diet in control mice (homocysteine 129 µmol/L), which was further worsened in T2DM db/db mice (homocysteine 180 µmol/L) with aggravated insulin intolerance. Hyperhomocysteinemia potentiated T2DM-induced mononuclear cell, monocyte, inflammatory monocyte (CD11b+Ly6C+), and M1 macrophage differentiation in periphery and aorta, which were rescued by folic acid-based homocysteine-lowering therapy. Moreover, hyperhomocysteinemia exacerbated T2DM-impaired endothelial-dependent aortic relaxation to acetylcholine. Finally, transfusion of bone marrow cells depleted for Ly6C by Ly6c shRNA transduction improved insulin intolerance and endothelial-dependent aortic relaxation in hyperhomocysteinemia+T2DM mice. CONCLUSIONS Hyperhomocysteinemia potentiated systemic and vessel wall inflammation and vascular dysfunction partially via inflammatory monocyte subset induction in T2DM. Inflammatory monocyte may be a novel therapeutic target for insulin resistance, inflammation, and cardiovascular complications in hyperhomocysteinemia+T2DM.
Collapse
Affiliation(s)
- Pu Fang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia PA
| | - Huimin Shan
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Jason J Saredy
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Ramon Cueto
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Jixiang Xia
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Xiaohua Jiang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
- Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
- Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
195
|
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
|
196
|
Polychronopoulou E, Braconnier P, Burnier M. New Insights on the Role of Sodium in the Physiological Regulation of Blood Pressure and Development of Hypertension. Front Cardiovasc Med 2019; 6:136. [PMID: 31608291 PMCID: PMC6756190 DOI: 10.3389/fcvm.2019.00136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/29/2019] [Indexed: 01/08/2023] Open
Abstract
A precise maintenance of sodium and fluid balance is an essential step in the regulation of blood pressure and alterations of this balance may lead to the development of hypertension. In recent years, several new advances were made in our understanding of the interaction between sodium and blood pressure regulation. The first is the discovery made possible with by new technology, such as 23Na-MRI, that sodium can be stored non-osmotically in tissues including the skin and muscles particularly when subjects are on a high sodium diet or have a reduced renal capacity to excrete sodium. These observations prompted the refinement of the original model of regulation of sodium balance from a two-compartment model comprising the extracellular fluid within the intravascular and interstitial spaces to a three-compartment model that includes the intracellular space of some tissues, most prominently the skin. In this new model, the immune system plays a role, thereby supporting many previous studies indicating that the immune system is a crucial co-contributor to the maintenance of hypertension through pro-hypertensive effects in the kidney, vasculature, and brain. Lastly, there is now evidence that sodium can affect the gut microbiome, and induce pro-inflammatory and immune responses, which might contribute to the development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Erietta Polychronopoulou
- Service of Nephrology and Hypertension, Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Philippe Braconnier
- Service of Nephrology and Hypertension, Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Michel Burnier
- Service of Nephrology and Hypertension, Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
197
|
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: 482] [Impact Index Per Article: 80.3] [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
|
198
|
Carnagarin R, Lambert GW, Kiuchi MG, Nolde JM, Matthews VB, Eikelis N, Lambert EA, Schlaich MP. Effects of sympathetic modulation in metabolic disease. Ann N Y Acad Sci 2019; 1454:80-89. [DOI: 10.1111/nyas.14217] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/07/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital Unit/Medical Research FoundationUniversity of Western Australia Perth Western Australia Australia
| | - Gavin W. Lambert
- Iverson Health Innovation Research InstituteSwinburne University of Technology Hawthorn Victoria Australia
- School of Health SciencesSwinburne University of Technology Hawthorn Victoria Australia
| | - Marcio G. Kiuchi
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital Unit/Medical Research FoundationUniversity of Western Australia Perth Western Australia Australia
| | - Janis M. Nolde
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital Unit/Medical Research FoundationUniversity of Western Australia Perth Western Australia Australia
| | - Vance B. Matthews
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital Unit/Medical Research FoundationUniversity of Western Australia Perth Western Australia Australia
| | - Nina Eikelis
- Iverson Health Innovation Research InstituteSwinburne University of Technology Hawthorn Victoria Australia
- School of Health SciencesSwinburne University of Technology Hawthorn Victoria Australia
| | - Elisabeth A. Lambert
- Iverson Health Innovation Research InstituteSwinburne University of Technology Hawthorn Victoria Australia
- School of Health SciencesSwinburne University of Technology Hawthorn Victoria Australia
| | - Markus P. Schlaich
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital Unit/Medical Research FoundationUniversity of Western Australia Perth Western Australia Australia
- Departments of Cardiology and NephrologyRoyal Perth Hospital Perth Western Australia Australia
- Neurovascular Hypertension and Kidney Disease LaboratoryBaker Heart and Diabetes Institute Melbourne Victoria Australia
| |
Collapse
|
199
|
Hilderman M, Qureshi AR, Abtahi F, Witt N, Jägren C, Olbers J, Delle M, Lindecrantz K, Bruchfeld A. The cholinergic anti-inflammatory pathway in resistant hypertension treated with renal denervation. Mol Med 2019; 25:39. [PMID: 31416428 PMCID: PMC6694612 DOI: 10.1186/s10020-019-0097-y] [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: 01/21/2019] [Accepted: 06/02/2019] [Indexed: 12/14/2022] Open
Abstract
Background Renal denervation (RDN) reduces sympathetic tone and may alter the sympathetic-parasympathetic balance. The autonomic nervous system is partly a regulator of innate immunity via the cholinergic anti-inflammatory pathway (CAP) which inhibits inflammation via the vagus nerve. Placental Growth Factor (PlGF) influences a neuro-immunological pathway in the spleen which may contribute to hypertension. The aim of this study was to investigate if modulation of renal sympathetic nerve activity affects CAP in terms of cytokine release as well as levels of PlGF. Methods Ten patients treated with RDN (Medtronic Inc), were analyzed for TNF, IL-1b and IL-10 and Lipopolysaccharide (LPS)-stimulated cytokine release before RDN, 1 day after and at 3- and 6-months follow-up. Four patients who underwent elective coronary angiography served as disease controls (DC). Results Baseline TNF was significantly lower 1 day after RDN (p = 0.03). LPS-stimulated (0, 10 and 100 ng/mL) TNF and IL-1b were significantly lower 1 day after RDN (TNF p = 0.0009, p = 0.0009 and p = 0.001, IL-1b; p = 0.0001, p = 0.002 and p = 0.005). IL-10 was significantly higher one day after RDN (p = ns, p = 0.02 and p = 0.01). These differences however declined during follow up. A more marked TNF reduction was achieved with a cholinergic analogue, GTS-21, in LPS-stimulated whole blood as compared with samples without GTS-21. Cytokine levels in controls did not differ before and 1 day after coronary angiography. PlGF was significantly higher in RDN patients and DC compared with healthy controls but did not change during follow-up. Conclusion RDN has an immediate effect on TNF in vivo and cytokine release ex vivo but seems to wane over time suggesting that current RDN techniques may not have long-lasting immunomodulatory effect. Repeated and extended stimulation of CAP in resistant hypertension by targeting neural circuits may be a potential therapeutic strategy for treatment of both hypertension and inflammation. Electronic supplementary material The online version of this article (10.1186/s10020-019-0097-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marie Hilderman
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Abdul Rashid Qureshi
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Farhad Abtahi
- Institute of Environmental Medicine, Division of Occupational Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nils Witt
- Department of Clinical Science and Education, Division of Cardiology, Karolinska Insititutet, Södersjukhuset, Stockholm, Sweden
| | - Christina Jägren
- Department of Clinical Science and Education, Division of Cardiology, Karolinska Insititutet, Södersjukhuset, Stockholm, Sweden
| | - Joakim Olbers
- Department of Clinical Science and Education, Division of Cardiology, Karolinska Insititutet, Södersjukhuset, Stockholm, Sweden
| | - Martin Delle
- Department of Radiology, Interventional Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Kaj Lindecrantz
- Institute of Environmental Medicine, Division of Occupational Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
200
|
Budzyń M, Gryszczyńka B, Boruczkowski M, Kaczmarek M, Begier-Krasińska B, Osińska A, Bukowska A, Iskra M, Kasprzak MP. The Potential Role of Circulating Endothelial Cells and Endothelial Progenitor Cells in the Prediction of Left Ventricular Hypertrophy in Hypertensive Patients. Front Physiol 2019; 10:1005. [PMID: 31447695 PMCID: PMC6696897 DOI: 10.3389/fphys.2019.01005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/19/2019] [Indexed: 01/20/2023] Open
Abstract
Background The main aim of present study is to evaluate the potential role of circulating endothelial cells (CECs) and endothelial progenitor cells (CEPCs) – representing specific markers of endothelial damage, in the prediction of left ventricular hypertrophy (LVH) in hypertensive patients categorized into two groups; mild (MH) and resistant hypertension (RH). Materials and Methods Thirty patients with MH and 28 subjects with RH were involved in the study. In both groups, patients were divided into an LVH and non-LVH group. The control group included 33 age and sex-matched normotensive volunteers. Physical examination, laboratory tests and echocardiography were conducted. Results In both the MH and RH group, patients with as well as without LVH demonstrated a higher number of CECs and a lower ratio of CEPCs/CECs as compared to the healthy control. Multiple linear regression analysis showed a positive association of CEPCs with left ventricular mass (LVM) and left ventricular mass index (LVMI), independently of other confounders. Conclusion Our results suggest that endothelial injury observed as an elevated CECs number and its impaired regeneration, reflected by a lowered CEPCs/CECs ratio, precede LVH occurrence and may play a significant role in LVH development regardless of the clinical severity of hypertension. Moreover, independent correlation of CEPCs with echocardiographic (ECG) incidences of LVH suggests their potential use as a screening biomarker to stratify the risk of LVH development.
Collapse
Affiliation(s)
- Magdalena Budzyń
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznań University of Medical Sciences, Poznań, Poland
| | - Bogna Gryszczyńka
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznań University of Medical Sciences, Poznań, Poland
| | - Maciej Boruczkowski
- Department of Clinical Immunology, Poznań University of Medical Sciences, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Clinical Immunology, Poznań University of Medical Sciences, Poznań, Poland
| | - Beata Begier-Krasińska
- Department of Hypertensiology, Angiology, and Internal Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Angelika Osińska
- Department of Hypertensiology, Angiology, and Internal Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Alicja Bukowska
- Medical Analysis Laboratory, Regional Blood Center, Poznań, Poland
| | - Maria Iskra
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznań University of Medical Sciences, Poznań, Poland
| | - Magdalena Paulina Kasprzak
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|