1
|
Kuntic M, Hahad O, Al-Kindi S, Oelze M, Lelieveld J, Daiber A, Münzel T. Pathomechanistic Synergy Between Particulate Matter and Traffic Noise-Induced Cardiovascular Damage and the Classical Risk Factor Hypertension. Antioxid Redox Signal 2024. [PMID: 38874533 DOI: 10.1089/ars.2024.0659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Affiliation(s)
- Marin Kuntic
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Omar Hahad
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Sadeer Al-Kindi
- Cardiovascular Prevention & Wellness and Center for CV Computational & Precision Health, Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Matthias Oelze
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jos Lelieveld
- Max Planck Institute for Chemistry, Atmospheric Chemistry, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| |
Collapse
|
2
|
Sørensen M, Pershagen G, Thacher JD, Lanki T, Wicki B, Röösli M, Vienneau D, Cantuaria ML, Schmidt JH, Aasvang GM, Al-Kindi S, Osborne MT, Wenzel P, Sastre J, Fleming I, Schulz R, Hahad O, Kuntic M, Zielonka J, Sies H, Grune T, Frenis K, Münzel T, Daiber A. Health position paper and redox perspectives - Disease burden by transportation noise. Redox Biol 2024; 69:102995. [PMID: 38142584 PMCID: PMC10788624 DOI: 10.1016/j.redox.2023.102995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023] Open
Abstract
Transportation noise is a ubiquitous urban exposure. In 2018, the World Health Organization concluded that chronic exposure to road traffic noise is a risk factor for ischemic heart disease. In contrast, they concluded that the quality of evidence for a link to other diseases was very low to moderate. Since then, several studies on the impact of noise on various diseases have been published. Also, studies investigating the mechanistic pathways underlying noise-induced health effects are emerging. We review the current evidence regarding effects of noise on health and the related disease-mechanisms. Several high-quality cohort studies consistently found road traffic noise to be associated with a higher risk of ischemic heart disease, heart failure, diabetes, and all-cause mortality. Furthermore, recent studies have indicated that road traffic and railway noise may increase the risk of diseases not commonly investigated in an environmental noise context, including breast cancer, dementia, and tinnitus. The harmful effects of noise are related to activation of a physiological stress response and nighttime sleep disturbance. Oxidative stress and inflammation downstream of stress hormone signaling and dysregulated circadian rhythms are identified as major disease-relevant pathomechanistic drivers. We discuss the role of reactive oxygen species and present results from antioxidant interventions. Lastly, we provide an overview of oxidative stress markers and adverse redox processes reported for noise-exposed animals and humans. This position paper summarizes all available epidemiological, clinical, and preclinical evidence of transportation noise as an important environmental risk factor for public health and discusses its implications on the population level.
Collapse
Affiliation(s)
- Mette Sørensen
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Denmark.
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jesse Daniel Thacher
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Timo Lanki
- Department of Health Security, Finnish Institute for Health and Welfare, Kuopio, Finland; School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Benedikt Wicki
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Danielle Vienneau
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Manuella Lech Cantuaria
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Jesper Hvass Schmidt
- Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Gunn Marit Aasvang
- Department of Air Quality and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Sadeer Al-Kindi
- Department of Medicine, University Hospitals, Harrington Heart & Vascular Institute, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Michael T Osborne
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Wenzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt Am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35392, Gießen, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Katie Frenis
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
3
|
Li H, Gao Y, Lin Y. Progress in molecular mechanisms of coronary microvascular dysfunction. Microcirculation 2023; 30:e12827. [PMID: 37608689 DOI: 10.1111/micc.12827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/23/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Coronary microvascular dysfunction is a high-risk factor for many cardiovascular events. However, because of multiple risk factors and limited understanding about its underlying pathophysiological mechanisms, it was easily misdiagnosed. Therefore, its clinical diagnosis and treatment were greatly restricted. Coronary microcirculation refers to microvessels that play an important role in the physiological regulation of myocardial perfusion and regulating blood flow distribution, fulfilling myocardial metabolic needs and moderating peripheral vascular resistance. In coronary microvascular dysfunction, vascular endothelial celldamage is a critical link. The main feature of early coronary microvascular dysfunction is the impairment of endothelial cell proliferation, adhesion, migration, apoptosis, and secretion. Moreover, coronary microvascular dysfunction risk factors include hyperglycemia, lipid metabolism disorders, ischemia-reperfusion injury, aging, and hypertension, similar to coronary atherosclerosis. There are various mechanisms by which these risk factors harm endothelial function and cause microcirculatory disturbances. Therefore, we reviewed coronary microvascular dysfunction's risk factors and pathogenesis in this article.
Collapse
Affiliation(s)
- Hao Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
4
|
Banecki KMRM, Dora KA. Endothelin-1 in Health and Disease. Int J Mol Sci 2023; 24:11295. [PMID: 37511055 PMCID: PMC10379484 DOI: 10.3390/ijms241411295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Discovered almost 40 years ago, the potent vasoconstrictor peptide endothelin-1 (ET-1) has a wide range of roles both physiologically and pathologically. In recent years, there has been a focus on the contribution of ET-1 to disease. This has led to the development of various ET receptor antagonists, some of which are approved for the treatment of pulmonary arterial hypertension, while clinical trials for other diseases have been numerous yet, for the most part, unsuccessful. However, given the vast physiological impact of ET-1, it is both surprising and disappointing that therapeutics targeting the ET-1 pathway remain limited. Strategies aimed at the pathways influencing the synthesis and release of ET-1 could provide new therapeutic avenues, yet research using cultured cells in vitro has had little follow up in intact ex vivo and in vivo preparations. This article summarises what is currently known about the synthesis, storage and release of ET-1 as well as the role of ET-1 in several diseases including cardiovascular diseases, COVID-19 and chronic pain. Unravelling the ET-1 pathway and identifying therapeutic targets has the potential to treat many diseases whether through disease prevention, slowing disease progression or reversing pathology.
Collapse
Affiliation(s)
| | - Kim A Dora
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
5
|
Daiber A, Kuntic M, Oelze M, Hahad O, Münzel T. E-cigarette effects on vascular function in animals and humans. Pflugers Arch 2023:10.1007/s00424-023-02813-z. [PMID: 37084087 DOI: 10.1007/s00424-023-02813-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/22/2023]
Abstract
Smoking tobacco cigarettes is a significant (cardiovascular) health risk factor. Although the number of tobacco cigarette users declined over the last decades, shisha smoking and e-cigarette vaping partially compensated for this health benefit. E-cigarettes may create highly addicted dual users (vaping and smoking). E-cigarettes seem not to represent a healthier alternative to tobacco smoking, although they may be less harmful. E-cigarette vaping causes oxidative stress, inflammation, endothelial dysfunction, and associated cardiovascular sequelae. This is primarily due to a significant overlap of toxic compounds in the vapor compared to tobacco smoke and, accordingly, a substantial overlap of pathomechanistic features between vaping and smoking. Whereas the main toxins in vapor are reactive aldehydes such as formaldehyde and acrolein, the toxic mixture in smoke is more complex, comprising particulate matter, reactive gases, transition metals, volatile organic compounds, and N-nitrosamines. However, it seems that both lifestyle drugs impair endothelial function to a quite similar extent, which may be due to the role of oxidative stress as the central pathomechanism to mediate endothelial dysfunction and vascular damage. Finally, the main selling argument for e-cigarette use that they help to quit smoking and get rid of nicotine addiction may be false because it seems that e-cigarettes instead trigger the opposite-younger entrance age and more frequent use. With our review, we summarize the adverse health impact of tobacco cigarettes and e-cigarettes, emphasizing the detrimental effects on endothelial function and cardiovascular health.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany.
| | - Marin Kuntic
- Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Matthias Oelze
- Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, Mainz, Germany.
| |
Collapse
|
6
|
Xu N, Zhu P, Yao Y, Jiang L, Jia S, Yuan D, Xu J, Wang H, Song Y, Gao L, Gao Z, Song L, Zhao X, Chen J, Yang Y, Xu B, Gao R, Yuan J. Big Endothelin-1 and long-term all-cause death in patients with coronary artery disease and prediabetes or diabetes after percutaneous coronary intervention. Nutr Metab Cardiovasc Dis 2022; 32:2147-2156. [PMID: 35843800 DOI: 10.1016/j.numecd.2022.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/15/2022] [Accepted: 06/04/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND AIMS The present study aimed to examine the association between big endothelin-1 (big ET-1) and long-term all-cause death in patients with coronary artery disease (CAD) and different glucose metabolism status. METHODS AND RESULTS We consecutively enrolled 8550 patients from January 2013 to December 2013. Patients were categorized according to both status of glucose metabolism status [Diabetes Mellitus (DM), Pre-Diabetes (Pre-DM), Normoglycemia (NG)] and big ET-1 levels. Primary endpoint was all-cause death. During a median of 5.1-year follow-up periods, 301 all-cause deaths occurred. Elevated big ET-1 was significantly associated with long-term all-cause death (adjusted HR: 2.230, 95%CI 1.629-3.051; p < 0.001). Similarly, patients with DM, but not Pre-DM, had increased risk of all-cause death compared with NG group (p < 0.05). When patients were categorized by both status of glucose metabolism and big ET-1 levels, high big ET-1 were associated with significantly higher risk of all-cause death in Pre-DM (adjusted HR: 2.442, 95% CI 1.039-5.740; p = 0.041) and DM (adjusted HR: 3.162, 95% CI 1.376-7.269; p = 0.007). The Kaplan-Meier curve indicated that DM patients with the highest big ET-1 levels were associated with the greatest risk of all-cause death (p < 0.05). CONCLUSIONS The present data indicate that baseline big ET-1 levels were independently associated with the long-term all-cause death in DM and Pre-DM patients with CAD undergoing PCI, suggesting that big ET-1 may be a valuable marker in patients with impaired glucose metabolism.
Collapse
Affiliation(s)
- Na Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Yao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Jiang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sida Jia
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deshan Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huanhuan Wang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lijian Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhan Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jilin Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runlin Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Posadino AM, Erre GL, Cossu A, Emanueli C, Eid AH, Zinellu A, Pintus G, Giordo R. NADPH-derived ROS generation drives fibrosis and endothelial-to-mesenchymal transition in systemic sclerosis: Potential cross talk with circulating miRNAs. Biomol Concepts 2022; 13:11-24. [PMID: 35189048 DOI: 10.1515/bmc-2021-0023] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis (SSc) is an immune disorder characterized by diffuse fibrosis and vascular abnormalities of the affected organs. Although the etiopathology of this disease is largely unknown, endothelial damage and oxidative stress appear implicated in its initiation and maintenance. Here, we show for the first time that circulating factors present in SSc sera increased reactive oxygen species (ROS) production, collagen synthesis, and proliferation of human pulmonary microvascular endothelial cells (HPMECs). The observed phenomena were also associated with endothelial to mesenchymal transition (EndMT) as indicated by decreased von Willebrand factor (vWF) expression and increased alpha-smooth muscle actin, respectively, an endothelial and mesenchymal marker. SSc-induced fibroproliferative effects were prevented by HPMECs exposition to the NADPH oxidase inhibitor diphenyleneiodonium, demonstrating ROS's causative role and suggesting their cellular origin. Sera from SSc patients showed significant changes in the expression of a set of fibrosis/EndMT-associated microRNAs (miRNA), including miR-21, miR-92a, miR-24, miR-27b, miR-125b, miR-29c, and miR-181b, which resulted significantly upregulated as compared to healthy donors sera. However, miR29b resulted downregulated in SSc sera, whereas no significant differences were found in the expression of miR-29a in the two experimental groups of samples. Taking together our data indicate NADPH oxidase-induced EndMT as a potential mechanism of SSc-associated fibrosis, suggesting fibrosis-associated miRNAs as potentially responsible for initiating and sustaining the vascular alterations observed in this pathological condition.
Collapse
Affiliation(s)
- Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, 07100 Sassari, Italy
| | - Annalisa Cossu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London, W12 0NN England, United Kingdom
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University Health, Qatar University, Doha, 2713, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, 2713, Qatar
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah 27272, United Arab Emirates
| | - Roberta Giordo
- Department of Basic Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| |
Collapse
|
8
|
Frenis K, Kuntic M, Hahad O, Bayo Jimenez MT, Oelze M, Daub S, Steven S, Münzel T, Daiber A. Redox Switches in Noise-Induced Cardiovascular and Neuronal Dysregulation. Front Mol Biosci 2021; 8:784910. [PMID: 34869603 PMCID: PMC8637611 DOI: 10.3389/fmolb.2021.784910] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Environmental exposures represent a significant health hazard, which cumulatively may be responsible for up to 2/3 of all chronic non-communicable disease and associated mortality (Global Burden of Disease Study and The Lancet Commission on Pollution and Health), which has given rise to a new concept of the exposome: the sum of environmental factors in every individual’s experience. Noise is part of the exposome and is increasingly being investigated as a health risk factor impacting neurological, cardiometabolic, endocrine, and immune health. Beyond the well-characterized effects of high-intensity noise on cochlear damage, noise is relatively well-studied in the cardiovascular field, where evidence is emerging from both human and translational experiments that noise from traffic-related sources could represent a risk factor for hypertension, ischemic heart disease, diabetes, and atherosclerosis. In the present review, we comprehensively discuss the current state of knowledge in the field of noise research. We give a brief survey of the literature documenting experiments in noise exposure in both humans and animals with a focus on cardiovascular disease. We also discuss the mechanisms that have been uncovered in recent years that describe how exposure to noise affects physiological homeostasis, leading to aberrant redox signaling resulting in metabolic and immune consequences, both of which have considerable impact on cardiovascular health. Additionally, we discuss the molecular pathways of redox involvement in the stress responses to noise and how they manifest in disruptions of the circadian rhythm, inflammatory signaling, gut microbiome composition, epigenetic landscape and vessel function.
Collapse
Affiliation(s)
- Katie Frenis
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Marin Kuntic
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | | | - Matthias Oelze
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Steffen Daub
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
9
|
The Causal Relationship between Endothelin-1 and Hypertension: Focusing on Endothelial Dysfunction, Arterial Stiffness, Vascular Remodeling, and Blood Pressure Regulation. Life (Basel) 2021; 11:life11090986. [PMID: 34575135 PMCID: PMC8472034 DOI: 10.3390/life11090986] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/01/2022] Open
Abstract
Hypertension (HTN) is one of the most prevalent diseases worldwide and is among the most important risk factors for cardiovascular and cerebrovascular complications. It is currently thought to be the result of disturbances in a number of neural, renal, hormonal, and vascular mechanisms regulating blood pressure (BP), so crucial importance is given to the imbalance of a number of vasoactive factors produced by the endothelium. Decreased nitric oxide production and increased production of endothelin-1 (ET-1) in the vascular wall may promote oxidative stress and low-grade inflammation, with the development of endothelial dysfunction (ED) and increased vasoconstrictor activity. Increased ET-1 production can contribute to arterial aging and the development of atherosclerotic changes, which are associated with increased arterial stiffness and manifestation of isolated systolic HTN. In addition, ET-1 is involved in the complex regulation of BP through synergistic interactions with angiotensin II, regulates the production of catecholamines and sympathetic activity, affects renal hemodynamics and water–salt balance, and regulates baroreceptor activity and myocardial contractility. This review focuses on the relationship between ET-1 and HTN and in particular on the key role of ET-1 in the pathogenesis of ED, arterial structural changes, and impaired vascular regulation of BP. The information presented includes basic concepts on the role of ET-1 in the pathogenesis of HTN without going into detailed analyses, which allows it to be used by a wide range of specialists. Also, the main pathological processes and mechanisms are richly illustrated for better understanding.
Collapse
|
10
|
Torres Crigna A, Link B, Samec M, Giordano FA, Kubatka P, Golubnitschaja O. Endothelin-1 axes in the framework of predictive, preventive and personalised (3P) medicine. EPMA J 2021; 12:265-305. [PMID: 34367381 PMCID: PMC8334338 DOI: 10.1007/s13167-021-00248-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
Endothelin-1 (ET-1) is involved in the regulation of a myriad of processes highly relevant for physical and mental well-being; female and male health; in the modulation of senses, pain, stress reactions and drug sensitivity as well as healing processes, amongst others. Shifted ET-1 homeostasis may influence and predict the development and progression of suboptimal health conditions, metabolic impairments with cascading complications, ageing and related pathologies, cardiovascular diseases, neurodegenerative pathologies, aggressive malignancies, modulating, therefore, individual outcomes of both non-communicable and infectious diseases such as COVID-19. This article provides an in-depth analysis of the involvement of ET-1 and related regulatory pathways in physiological and pathophysiological processes and estimates its capacity as a predictor of ageing and related pathologies,a sensor of lifestyle quality and progression of suboptimal health conditions to diseases for their targeted preventionand as a potent target for cost-effective treatments tailored to the person.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Barbara Link
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
11
|
Münzel T, Templin C, Cammann VL, Hahad O. Takotsubo Syndrome: Impact of endothelial dysfunction and oxidative stress. Free Radic Biol Med 2021; 169:216-223. [PMID: 33864955 DOI: 10.1016/j.freeradbiomed.2021.03.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 12/30/2022]
Abstract
Takotsubo Syndrome (TTS) is characterized by a transient left ventricular dysfunction recovering spontaneously within days or weeks. Although the pathophysiology of TTS remains obscure, there is growing evidence suggesting TTS to be associated with increased production of reactive oxygen species (ROS), which may be involved in causing transient coronary and peripheral endothelial dysfunction leading to a transient impairment of myocardial contraction due to stunning (apical ballooning). Endothelial dysfunction is mainly caused by decreased vascular and myocardial nitric oxide bioavailability in response to increased ROS production. Accordingly, studies in humans and animal models demonstrated increased myocardial dihydroethidium staining of the myocardium in endomyocardial biopsy specimens, increased levels of hydrogen peroxide and malondialdehyde as well as reduced glutathione levels compatible with increased oxidative stress. As significant superoxide sources the mitochondria and the NADPH oxidase isoform NOX-4 and the NOX-2 regulating cytosolic subunit p67phox have been identified. Treatment with antioxidants such as sodium hydrosulfide reduced superoxide production in mitochondria and reduced expression of NOX-4 and p67phox, respectively. The presence of superoxide and nitric oxide also provides the basis for the concept of nitro-oxidative as well as nitrosative stress in TTS.
Collapse
Affiliation(s)
- Thomas Münzel
- Department of Cardiology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany.
| | | | | | - Omar Hahad
- Department of Cardiology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
12
|
Kuntic M, Oelze M, Steven S, Kröller-Schön S, Stamm P, Kalinovic S, Frenis K, Vujacic-Mirski K, Bayo Jimenez MT, Kvandova M, Filippou K, Al Zuabi A, Brückl V, Hahad O, Daub S, Varveri F, Gori T, Huesmann R, Hoffmann T, Schmidt FP, Keaney JF, Daiber A, Münzel T. Short-term e-cigarette vapour exposure causes vascular oxidative stress and dysfunction: evidence for a close connection to brain damage and a key role of the phagocytic NADPH oxidase (NOX-2). Eur Heart J 2021; 41:2472-2483. [PMID: 31715629 PMCID: PMC7340357 DOI: 10.1093/eurheartj/ehz772] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/12/2019] [Accepted: 10/19/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS Electronic (e)-cigarettes have been marketed as a 'healthy' alternative to traditional combustible cigarettes and as an effective method of smoking cessation. There are, however, a paucity of data to support these claims. In fact, e-cigarettes are implicated in endothelial dysfunction and oxidative stress in the vasculature and the lungs. The mechanisms underlying these side effects remain unclear. Here, we investigated the effects of e-cigarette vapour on vascular function in smokers and experimental animals to determine the underlying mechanisms. METHODS AND RESULTS Acute e-cigarette smoking produced a marked impairment of endothelial function in chronic smokers determined by flow-mediated dilation. In mice, e-cigarette vapour without nicotine had more detrimental effects on endothelial function, markers of oxidative stress, inflammation, and lipid peroxidation than vapour containing nicotine. These effects of e-cigarette vapour were largely absent in mice lacking phagocytic NADPH oxidase (NOX-2) or upon treatment with the endothelin receptor blocker macitentan or the FOXO3 activator bepridil. We also established that the e-cigarette product acrolein, a reactive aldehyde, recapitulated many of the NOX-2-dependent effects of e-cigarette vapour using in vitro blood vessel incubation. CONCLUSIONS E-cigarette vapour exposure increases vascular, cerebral, and pulmonary oxidative stress via a NOX-2-dependent mechanism. Our study identifies the toxic aldehyde acrolein as a key mediator of the observed adverse vascular consequences. Thus, e-cigarettes have the potential to induce marked adverse cardiovascular, pulmonary, and cerebrovascular consequences. Since e-cigarette use is increasing, particularly amongst youth, our data suggest that aggressive steps are warranted to limit their health risks.
Collapse
Affiliation(s)
- Marin Kuntic
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Sebastian Steven
- Center for Cardiology, University Medical Center, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center, Mainz, German
| | | | - Paul Stamm
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Sanela Kalinovic
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Katie Frenis
- Center for Cardiology, University Medical Center, Mainz, Germany
| | | | | | | | | | - Ahmad Al Zuabi
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Vivienne Brückl
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Omar Hahad
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Steffen Daub
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Franco Varveri
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - Tommaso Gori
- Center for Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Regina Huesmann
- Institute for Inorganic and Analytical Chemistry, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thorsten Hoffmann
- Institute for Inorganic and Analytical Chemistry, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Frank P Schmidt
- Center for Cardiology, University Medical Center, Mainz, Germany
| | - John F Keaney
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Andreas Daiber
- Center for Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Thomas Münzel
- Center for Cardiology, University Medical Center, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center, Mainz, German.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| |
Collapse
|
13
|
de Oliveira AA, Nunes KP. Hypertension and Erectile Dysfunction: Breaking Down the Challenges. Am J Hypertens 2021; 34:134-142. [PMID: 32866225 DOI: 10.1093/ajh/hpaa143] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/15/2020] [Accepted: 08/29/2020] [Indexed: 02/07/2023] Open
Abstract
A diagnostic of hypertension increases the risk of erectile dysfunction (ED); likewise, ED can be an early sign of hypertension. In both cases, there is evidence that endothelial dysfunction is a common link between the 2 conditions. During hypertension, the sustained and widespread release of procontractile factors (e.g., angiotensin II, endothelin 1, and aldosterone) impairs the balance between vasoconstrictors and vasodilators and, in turn, detrimentally impacts vascular and erectile structures. This prohypertensive state associates with an enhancement in the generation of reactive oxygen species, which is not compensated by internal antioxidant mechanisms. Recently, the innate immune system, mainly via Toll-like receptor 4, has also been shown to actively contribute to the pathophysiology of hypertension and ED not only by inducing oxidative stress but also by sustaining a low-grade inflammatory state. Furthermore, some drugs used to treat hypertension can cause ED and, consequently, reduce compliance with the prescribed pharmacotherapy. To break down these challenges, in this review, we focus on discussing the well-established as well as the emerging mechanisms linking hypertension and ED with an emphasis on the signaling network of the vasculature and corpora cavernosa, the vascular-like structure of the penis.
Collapse
Affiliation(s)
- Amanda Almeida de Oliveira
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida, USA
| | - Kenia Pedrosa Nunes
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida, USA
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Daiber A, Kröller-Schön S, Oelze M, Hahad O, Li H, Schulz R, Steven S, Münzel T. Oxidative stress and inflammation contribute to traffic noise-induced vascular and cerebral dysfunction via uncoupling of nitric oxide synthases. Redox Biol 2020; 34:101506. [PMID: 32371009 PMCID: PMC7327966 DOI: 10.1016/j.redox.2020.101506] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Environmental pollution and non-chemical stressors such as mental stress or traffic noise exposure are increasingly accepted as health risk factors with substantial contribution to chronic noncommunicable diseases (e.g. cardiovascular, metabolic and mental). Whereas the mechanisms of air pollution-mediated adverse health effects are well characterized, the mechanisms of traffic noise exposure are not completely understood, despite convincing clinical and epidemiological evidence for a significant contribution of environmental noise to overall mortality and disability. The initial mechanism of noise-induced cardiovascular, metabolic and mental disease is well defined by the „noise reaction model“ and consists of neuronal activation involving the hypothalamic-pituitary-adrenal (HPA) axis as well as the sympathetic nervous system, followed by a classical stress response via cortisol and catecholamines. Stress pathways are initiated by noise-induced annoyance and sleep deprivation/fragmentation. This review highlights the down-stream pathophysiology of noise-induced mental stress, which is based on an induction of inflammation and oxidative stress. We highlight the sources of reactive oxygen species (ROS) involved and the known targets for noise-induced oxidative damage. Part of the review emphasizes noise-triggered uncoupling/dysregulation of endothelial and neuronal nitric oxide synthase (eNOS and nNOS) and its central role for vascular dysfunction. Exposure to (traffic) noise causes non-auditory (indirect) cardiovascular and cerebral health harms via neuronal activation. Noise activates the HPA axis and sympathetic nervous system increasing levels of stress hormones, vasoconstrictors and ROS. Noise induces inflammation and stimulates several ROS sources leading to cerebral and cardiovascular oxidative damage. Noise leads to eNOS and nNOS uncoupling contributing to cardiometabolic disease and cognitive impairment.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Swenja Kröller-Schön
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Omar Hahad
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Mainz, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Sebastian Steven
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
17
|
Mitchell T, De Miguel C, Gohar EY. Sex differences in redox homeostasis in renal disease. Redox Biol 2020; 31:101489. [PMID: 32197946 PMCID: PMC7212488 DOI: 10.1016/j.redox.2020.101489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/20/2020] [Accepted: 03/01/2020] [Indexed: 02/08/2023] Open
Abstract
Sex differences in redox signaling in the kidney present new challenges and opportunities for understanding the physiology and pathophysiology of the kidney. This review will focus on reactive oxygen species, immune-related signaling pathways and endothelin-1 as potential mediators of sex-differences in redox homeostasis in the kidney. Additionally, this review will highlight male-female differences in redox signaling in several major cardiovascular and renal disorders namely acute kidney injury, diabetic nephropathy, kidney stone disease and salt-sensitive hypertension. Furthermore, we will discuss the contribution of redox signaling in the pathogenesis of postmenopausal hypertension and preeclampsia.
Collapse
Affiliation(s)
- Tanecia Mitchell
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eman Y Gohar
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
18
|
Abstract
The microcirculation maintains tissue homeostasis through local regulation of blood flow and oxygen delivery. Perturbations in microvascular function are characteristic of several diseases and may be early indicators of pathological changes in the cardiovascular system and in parenchymal tissue function. These changes are often mediated by various reactive oxygen species and linked to disruptions in pathways such as vasodilation or angiogenesis. This overview compiles recent advances relating to redox regulation of the microcirculation by adopting both cellular and functional perspectives. Findings from a variety of vascular beds and models are integrated to describe common effects of different reactive species on microvascular function. Gaps in understanding and areas for further research are outlined. © 2020 American Physiological Society. Compr Physiol 10:229-260, 2020.
Collapse
Affiliation(s)
- Andrew O Kadlec
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - David D Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Medicine-Division of Cardiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
19
|
Grotle AK, Stone AJ. Exaggerated exercise pressor reflex in type 2 diabetes: Potential role of oxidative stress. Auton Neurosci 2019; 222:102591. [PMID: 31669797 PMCID: PMC6858935 DOI: 10.1016/j.autneu.2019.102591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) leads to exaggerated cardiovascular responses to exercise, in part due to an exaggerated exercise pressor reflex. Accumulating data suggest excessive oxidative stress contributes to an exaggerated exercise pressor reflex in cardiovascular-related diseases. Excessive oxidative stress is also a primary underlying mechanism for the development and progression of T2DM. However, whether oxidative stress plays a role in mediating the exaggerated exercise pressor reflex in T2DM is not known. Therefore, this review explores the potential role of oxidative stress leading to increased activation of the afferent arm of the exercise pressor reflex. Several lines of evidence support direct and indirect effects of oxidative stress on the exercise pressor reflex. For example, intramuscular ROS may directly and indirectly (by attenuating contracting muscle blood flow) increase group III and IV afferent activity. Oxidative stress is a primary underlying mechanism for the development of neuropathic pain, which in turn is associated with increased group III and IV afferent activity. These are the same type of afferents that evoke muscle pain and the exercise pressor reflex. Furthermore, oxidative stress-induced release of inflammatory mediators may modulate afferent activity. Collectively, these alterations may result in a positive feedback loop that further amplifies the exercise pressor reflex. An exaggerated reflex increases the risk of adverse cardiovascular events. Thus, identifying the contribution of oxidative stress could provide a potential therapeutic target to reduce this risk in T2DM.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Audrey J Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX 78712, United States of America.
| |
Collapse
|
20
|
Epstein JB, Miaskowski C. Oral Pain in the Cancer Patient. J Natl Cancer Inst Monogr 2019; 2019:5551353. [DOI: 10.1093/jncimonographs/lgz003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/07/2019] [Accepted: 04/01/2019] [Indexed: 01/02/2023] Open
Abstract
Abstract
Oral pain due to cancer and associated treatments is common. The prevalence and severity of oral cancer is high. Painful oral mucositis develops in head and neck cancer patients following surgery and associated radiation therapy and/or chemotherapy. In addition, oral pain, including pain from mucositis, occurs in patients receiving chemotherapy for cancers of the hematopoietic system and cancers at other anatomic sites. Despite pain management practices that include high-dose opioid analgesics, patients rarely obtain relief from either head and neck cancer pain or mucositis pain. Because oral pain in cancer patients is likely due to both nociceptive and neuropathic mechanisms, effective management of pain requires treatments for both processes. As knowledge of the pathophysiology of oral pain in cancer patients increases, new approaches for the prevention and management are anticipated. This article focuses on the emerging evidence that supports the molecular mechanisms and the unique oral micro-neuroanatomy that in combination produce the severe oral pain experienced by cancer patients. In addition, this article summarizes the current state of clinical management of oral mucositis pain.
Collapse
Affiliation(s)
- Joel B Epstein
- Department of Surgery, City of Hope, Duarte, CA
- Department of Surgery, Cedars-Sinai Health System, Los Angeles, CA
- Seattle Cancer Care Alliance, Seattle, WA
| | - Christine Miaskowski
- Department of Physiological Nursing, University of California San Francisco, San Francisco, CA
| |
Collapse
|
21
|
Yu H, Kalogeris T, Korthuis RJ. Reactive species-induced microvascular dysfunction in ischemia/reperfusion. Free Radic Biol Med 2019; 135:182-197. [PMID: 30849489 PMCID: PMC6503659 DOI: 10.1016/j.freeradbiomed.2019.02.031] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells line the inner surface of the entire cardiovascular system as a single layer and are involved in an impressive array of functions, ranging from the regulation of vascular tone in resistance arteries and arterioles, modulation of microvascular barrier function in capillaries and postcapillary venules, and control of proinflammatory and prothrombotic processes, which occur in all segments of the vascular tree but can be especially prominent in postcapillary venules. When tissues are subjected to ischemia/reperfusion (I/R), the endothelium of resistance arteries and arterioles, capillaries, and postcapillary venules become dysfunctional, resulting in impaired endothelium-dependent vasodilator and enhanced endothelium-dependent vasoconstrictor responses along with increased vulnerability to thrombus formation, enhanced fluid filtration and protein extravasation, and increased blood-to-interstitium trafficking of leukocytes in these functionally distinct segments of the microcirculation. The number of capillaries open to flow upon reperfusion also declines as a result of I/R, which impairs nutritive perfusion. All of these pathologic microvascular events involve the formation of reactive species (RS) derived from molecular oxygen and/or nitric oxide. In addition to these effects, I/R-induced RS activate NLRP3 inflammasomes, alter connexin/pannexin signaling, provoke mitochondrial fission, and cause release of microvesicles in endothelial cells, resulting in deranged function in arterioles, capillaries, and venules. It is now apparent that this microvascular dysfunction is an important determinant of the severity of injury sustained by parenchymal cells in ischemic tissues, as well as being predictive of clinical outcome after reperfusion therapy. On the other hand, RS production at signaling levels promotes ischemic angiogenesis, mediates flow-induced dilation in patients with coronary artery disease, and instigates the activation of cell survival programs by conditioning stimuli that render tissues resistant to the deleterious effects of prolonged I/R. These topics will be reviewed in this article.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ted Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA.
| |
Collapse
|
22
|
Gu Y, Liang Y, Bai J, Wu W, Lin Q, Wu J. Spent hen-derived ACE inhibitory peptide IWHHT shows antioxidative and anti-inflammatory activities in endothelial cells. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
23
|
The Endothelin Receptor Antagonist Macitentan Improves Isosorbide-5-Mononitrate (ISMN) and Isosorbide Dinitrate (ISDN) Induced Endothelial Dysfunction, Oxidative Stress, and Vascular Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2018:7845629. [PMID: 30687454 PMCID: PMC6327264 DOI: 10.1155/2018/7845629] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/17/2018] [Indexed: 11/18/2022]
Abstract
Objective Organic nitrates such as isosorbide-5-mononitrate (ISMN) and isosorbide dinitrate (ISDN) are used for the treatment of patients with chronic symptomatic stable coronary artery disease and chronic congestive heart failure. Limiting side effects of these nitrovasodilators include nitrate tolerance and/or endothelial dysfunction mediated by oxidative stress. Here, we tested the therapeutic effects of the dual endothelin (ET) receptor antagonist macitentan in ISMN- and ISDN-treated animals. Methods and Results Organic nitrates (ISMN, ISDN, and nitroglycerin (GTN)) augmented the oxidative burst and interleukin-6 release in cultured macrophages, whereas macitentan decreased the oxidative burst in isolated human leukocytes. Male C57BL/6j mice were treated with ISMN (75 mg/kg/d) or ISDN (25 mg/kg/d) via s.c. infusion for 7 days and some mice in addition with 30 mg/kg/d of macitentan (gavage, once daily). ISMN and ISDN in vivo therapy caused endothelial dysfunction but no nitrate (or cross-)tolerance to the organic nitrates, respectively. ISMN/ISDN increased blood nitrosative stress, vascular/cardiac oxidative stress via NOX-2 (fluorescence and chemiluminescence methods), ET1 expression, ET receptor signaling, and markers of inflammation (protein and mRNA level). ET receptor signaling blockade by macitentan normalized endothelial function, vascular/cardiac oxidative stress, and inflammatory phenotype in both nitrate therapy groups. Conclusion ISMN/ISDN treatment caused activation of the NOX-2/ET receptor signaling axis leading to increased vascular oxidative stress and inflammation as well as endothelial dysfunction. Our study demonstrates for the first time that blockade of ET receptor signaling by the dual endothelin receptor blocker macitentan improves adverse side effects of the organic nitrates ISMN and ISDN.
Collapse
|
24
|
Münzel T, Daiber A, Steven S, Tran LP, Ullmann E, Kossmann S, Schmidt FP, Oelze M, Xia N, Li H, Pinto A, Wild P, Pies K, Schmidt ER, Rapp S, Kröller-Schön S. Effects of noise on vascular function, oxidative stress, and inflammation: mechanistic insight from studies in mice. Eur Heart J 2018; 38:2838-2849. [PMID: 28329261 DOI: 10.1093/eurheartj/ehx081] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023] Open
Abstract
Aims Epidemiological studies indicate that traffic noise increases the incidence of coronary artery disease, hypertension and stroke. The underlying mechanisms remain largely unknown. Field studies with nighttime noise exposure demonstrate that aircraft noise leads to vascular dysfunction, which is markedly improved by vitamin C, suggesting a key role of oxidative stress in causing this phenomenon. Methods and results We developed a novel animal model to study the vascular consequences of aircraft noise exposure. Peak sound levels of 85 and mean sound level of 72 dBA applied by loudspeakers for 4 days caused an increase in systolic blood pressure, plasma noradrenaline and angiotensin II levels and induced endothelial dysfunction. Noise increased eNOS expression but reduced vascular NO levels because of eNOS uncoupling. Noise increased circulating levels of nitrotyrosine, interleukine-6 and vascular expression of the NADPH oxidase subunit Nox2, nitrotyrosine-positive proteins and of endothelin-1. FACS analysis demonstrated an increase in infiltrated natural killer-cells and neutrophils into the vasculature. Equal mean sound pressure levels of white noise for 4 days did not induce these changes. Comparative Illumina sequencing of transcriptomes of aortic tissues from aircraft noise-treated animals displayed significant changes of genes in part responsible for the regulation of vascular function, vascular remodelling, and cell death. Conclusion We established a novel and unique aircraft noise stress model with increased blood pressure and vascular dysfunction associated with oxidative stress. This animal model enables future studies of molecular mechanisms, mitigation strategies, and pharmacological interventions to protect from noise-induced vascular damage.
Collapse
Affiliation(s)
- Thomas Münzel
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Andreas Daiber
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Sebastian Steven
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Lan P Tran
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Elisabeth Ullmann
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Sabine Kossmann
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Frank P Schmidt
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Obere Zahlbacher , Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Obere Zahlbacher , Mainz, Germany
| | - Antonio Pinto
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp Wild
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kai Pies
- Engineering Office for Noise Protection, Mainz, Germany
| | - Erwin R Schmidt
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Steffen Rapp
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
25
|
Mandel ER, Dunford EC, Abdifarkosh G, Turnbull PC, Perry CGR, Riddell MC, Haas TL. The superoxide dismutase mimetic tempol does not alleviate glucocorticoid-mediated rarefaction of rat skeletal muscle capillaries. Physiol Rep 2018; 5:e13243. [PMID: 28533261 PMCID: PMC5449555 DOI: 10.14814/phy2.13243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 12/17/2022] Open
Abstract
Sustained elevations in circulating glucocorticoids elicit reductions in skeletal muscle microvascular content, but little is known of the underlying mechanisms. We hypothesized that glucocorticoid‐induced oxidative stress contributes to this phenomenon. In rats that were implanted with corticosterone (CORT) or control pellets, CORT caused a significant decrease in muscle glutathione levels and a corresponding increase in protein carbonylation, an irreversible oxidative modification of proteins. Decreased endothelial nitric oxide synthase and increased endothelin‐1 mRNA levels were detected after 9 days of CORT, and blood flow to glycolytic muscles was diminished. Control and CORT rats were treated concurrently with drinking water containing the superoxide dismutase mimetic tempol (172 mg/L) or the α‐1 adrenergic receptor antagonist prazosin (50 mg/L) for 6 or 16 days. Both tempol and prazosin alleviated skeletal muscle protein carbonylation. Tempol failed to prevent CORT‐mediated capillary rarefaction and was ineffective in restoring skeletal muscle blood flow. In contrast, prazosin blocked capillary rarefaction and restored skeletal muscle blood flow to control levels. The failure of tempol to prevent CORT‐induced skeletal muscle microvascular rarefaction does not support a dominant role of superoxide‐induced oxidative stress in this process. Although a decrease in protein carbonylation was observed with prazosin treatment, our data suggest that the maintenance of skeletal muscle microvascular content is related more closely with counteracting the CORT‐mediated influence on skeletal muscle vascular tone.
Collapse
Affiliation(s)
- Erin R Mandel
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Emily C Dunford
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Ghoncheh Abdifarkosh
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Patrick C Turnbull
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Michael C Riddell
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Tara L Haas
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Jurrissen TJ, Olver TD, Winn NC, Grunewald ZI, Lin GS, Hiemstra JA, Edwards JC, Gastecki ML, Welly RJ, Emter CA, Vieira-Potter, VJ, Padilla J. Endothelial dysfunction occurs independently of adipose tissue inflammation and insulin resistance in ovariectomized Yucatan miniature-swine. Adipocyte 2018; 7:35-44. [PMID: 29283284 DOI: 10.1080/21623945.2017.1405191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
In rodents, experimentally-induced ovarian hormone deficiency increases adiposity and adipose tissue (AT) inflammation, which is thought to contribute to insulin resistance and increased cardiovascular disease risk. However, whether this occurs in a translationally-relevant large animal model remains unknown. Herein, we tested the hypothesis that ovariectomy would promote visceral and perivascular AT (PVAT) inflammation, as well as subsequent insulin resistance and peripheral vascular dysfunction in female swine. At sexual maturity (7 months of age), female Yucatan mini-swine either remained intact (control, n = 9) or were ovariectomized (OVX, n = 7). All pigs were fed standard chow (15-20 g/kg), and were euthanized 6 months post-surgery. Uterine mass and plasma estradiol levels were decreased by ∼10-fold and 2-fold, respectively, in OVX compared to control pigs. Body mass, glucose homeostasis, and markers of insulin resistance were not different between control and OVX pigs; however, OVX animals exhibited greater plasma triglycerides and triglyceride:HDL ratio. Ovariectomy enhanced visceral adipocyte expansion, although this was not accompanied by brachial artery PVAT adipocyte expansion, AT inflammation in either depot, or increased systemic inflammation assessed by plasma C-reactive protein concentrations. Despite the lack of AT inflammation and insulin resistance, OVX pigs exhibited depressed brachial artery endothelial-dependent vasorelaxation, which was rescued with blockade of endothelin receptor A. Together, these findings indicate that in female Yucatan mini-swine, increased AT inflammation and insulin resistance are not required for loss of ovarian hormones to induce endothelial dysfunction.
Collapse
Affiliation(s)
- Thomas J. Jurrissen
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - T. Dylan Olver
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Nathan C. Winn
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Zachary I. Grunewald
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Gabriela S. Lin
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Biology, Barry University, Miami, FL, United States
| | | | - Jenna C. Edwards
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Michelle L. Gastecki
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Rebecca J. Welly
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Craig A. Emter
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | | | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Child Health, University of Missouri, Columbia, MO, United States
| |
Collapse
|
27
|
Saleh MA, De Miguel C, Stevens DI, Carmines PK, Pollock DM, Pollock JS. Free radical scavenging decreases endothelin-1 excretion and glomerular albumin permeability during type 1 diabetes. Physiol Rep 2017; 4:4/24/e13055. [PMID: 28039404 PMCID: PMC5210388 DOI: 10.14814/phy2.13055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 11/24/2022] Open
Abstract
Increased renal endothelin-1 (ET-1) production and an ETA receptor-dependent increase in glomerular albumin permeability (Palb) accompany type 1 diabetes mellitus (T1D). We hypothesized that T1D-induced oxidative stress contributes to renal ET-1 production and glomerular Palb Male rats with streptozotocin-induced T1D were provided free access to drinking water without additives (T1D rats) or containing the free radical scavenger tempol (1 mmol/L; T1D+Tempol). After 3 weeks, T1D+Tempol rats displayed lower urinary excretion of thiobarbituric acid reactive substances and glomerular superoxide production (dihydroethidium staining) compared to T1D rats. Urinary ET-1 excretion and inner medullary (but not cortical or outer medullary) prepro-ET-1 mRNA expression were lower in the T1D+Tempol group than in the T1D group. Palb, measured as the change in volume of isolated glomeruli upon exposure to oncotic gradients of albumin, was significantly lower in the T1D+Tempol group than in the T1D group. Tempol treatment did not alter protein excretion or creatinine clearance. These data support the postulate that oxidative stress contributes to glomerular Palb and renal ET-1 production during the early phase of type 1 diabetes.
Collapse
Affiliation(s)
- Mohamed A Saleh
- Medical College of Georgia, Augusta University, Augusta, Georgia.,Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Carmen De Miguel
- Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David I Stevens
- Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Pamela K Carmines
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - David M Pollock
- Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jennifer S Pollock
- Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama .,Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
28
|
Gurusinghe S, Cox AG, Rahman R, Chan ST, Muljadi R, Singh H, Leaw B, Mockler JC, Marshall SA, Murthi P, Lim R, Wallace EM. Resveratrol mitigates trophoblast and endothelial dysfunction partly via activation of nuclear factor erythroid 2-related factor-2. Placenta 2017; 60:74-85. [PMID: 29208243 DOI: 10.1016/j.placenta.2017.10.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/03/2017] [Accepted: 10/30/2017] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Maternal endothelial dysfunction underlying preeclampsia arises from excessive placental release of anti-angiogenic factors, such as soluble fms-like tyrosine kinase-1 (sFlt1), soluble endoglin (sEng) and activin A. Resveratrol, an activator of the nuclear factor erythroid 2-related factor-2 (Nrf2) transcription factor, mediates the gene expression of antioxidant and vasoprotective factors that may counter the endothelial damage imposed by these anti-angiogenic factors. The objective of this study was to assess whether resveratrol could reduce placental oxidative stress and production of anti-angiogenic factors in vitro and/or improve in vitro markers of endothelial dysfunction via Nrf2 activation. METHOD We used in vitro term placental explants to assess the effects of resveratrol on placental oxidative stress and production of sFlt1, sEng and activin A. Using human umbilical vein endothelial cells we investigated the effects of resveratrol on markers of in vitro endothelial dysfunction, including the expression of intercellular adhesion molecule 1 (ICAM1), vascular cell adhesion molecule 1 (VCAM1), E-selectin and endothelin-1, and endothelial permeability. To confirm that resveratrol mediated its effects via Nrf2, we examined the impact of resveratrol on the same in vitro markers of endothelial and placental dysfunction following Nrf2 knockdown. RESULTS Resveratrol significantly decreased placental oxidative stress and the production of sFlt1 and activin A. Resveratrol significantly mitigated tumor necrosis factor-α stimulated endothelial expression of ICAM1, VCAM1, E-selectin and endothelin-1 and prevented an increase in endothelial monolayer permeability. Nrf2 knockdown abolished some of the protective effects of resveratrol on endothelial cells, but not in primary trophoblast cells. CONCLUSION Features of placental and endothelial dysfunction characteristic of preeclampsia are improved by resveratrol in vitro, partially via the modulation of Nrf2.
Collapse
Affiliation(s)
- Seshini Gurusinghe
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Annie G Cox
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Rahana Rahman
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Siow T Chan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Ruth Muljadi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Harmeet Singh
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Bryan Leaw
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Joanne C Mockler
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Padma Murthi
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Euan M Wallace
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Monash Women's Services, Monash Health, Clayton, Victoria, Australia.
| |
Collapse
|
29
|
Wenzel P, Kossmann S, Münzel T, Daiber A. Redox regulation of cardiovascular inflammation - Immunomodulatory function of mitochondrial and Nox-derived reactive oxygen and nitrogen species. Free Radic Biol Med 2017; 109:48-60. [PMID: 28108279 DOI: 10.1016/j.freeradbiomed.2017.01.027] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/16/2017] [Indexed: 12/18/2022]
Abstract
Oxidative stress is a major hallmark of cardiovascular diseases although a causal link was so far not proven by large clinical trials. However, there is a close association between oxidative stress and inflammation and increasing evidence for a causal role of (low-grade) inflammation for the onset and progression of cardiovascular diseases, which may serve as the missing link between oxidative stress and cardiovascular morbidity and mortality. With the present review we would like to highlight the multiple redox regulated pathways in inflammation, discuss the sources of reactive oxygen and nitrogen species that are of interest for these processes and finally discuss the importance of angiotensin II (AT-II) as a trigger of cardiovascular inflammation and the initiation and progression of cardiovascular diseases.
Collapse
Affiliation(s)
- Philip Wenzel
- Center for Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Center of Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Sabine Kossmann
- Center for Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Center of Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Center of Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Mainz, Germany.
| |
Collapse
|
30
|
Wheatley W, Kohan DE. Role for reactive oxygen species in flow-stimulated inner medullary collecting duct endothelin-1 production. Am J Physiol Renal Physiol 2017; 313:F514-F521. [PMID: 28515175 PMCID: PMC5582894 DOI: 10.1152/ajprenal.00103.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/08/2017] [Accepted: 05/13/2017] [Indexed: 12/22/2022] Open
Abstract
Inner medullary collecting duct (IMCD)-derived endothelin-1 (ET-1) is stimulated by volume expansion, in part through augmented luminal flow, whereupon it can elicit natriuresis and diuresis. Since flow can alter nitric oxide (NO) and reactive oxygen species (ROS), both of which can affect collecting duct salt transport, we asked whether NO and/or ROS mediate flow-stimulated IMCD ET-1. Mouse IMCD3 cells were exposed to flow, and ET-1/GAPDH mRNA was assessed. A shear stress of 10 dyn/cm2 for 1 h increased ET-1 mRNA by fourfold compared with no flow (ET-1 flow response). Global NO synthase (NOS) inhibition [NG-nitro-l-arginine methyl ester (l-NAME)] reduced the ET-1 flow response; however, pharmacological inhibition of NOS1 or NOS2, inhibition of NOS3 siRNA, inhibition of arginase inhibition, removal of media l-Arg, or inhibition of NO-dependent signaling pathways (PKG, guanylyl cyclase, or NF-κB) did not affect the ET-1 flow response. Tempol reduced the ET-1 flow response; no further inhibition occurred with l-NAME. Superoxide dismutase, but not catalase, reduced the ET-1 flow response. Inhibition of NAPDH oxidase (NOX) (apocynin), pharmacological inhibition of NOX1/4, or NOX4 siRNA reduced the ET-1 flow response. Finally, flow increased IMCD3 ROS production and this was inhibited by apocynin, NOX1/4 inhibition, and, to a small extent, by l-NAME. Taken together, these data suggest that NOX4-derived ROS in general, and possibly superoxide in particular, are involved in flow-stimulated IMCD ET-1 production. To our knowledge, this is the first report of flow-stimulated ROS production by the CD, as well as the first report of such flow-stimulated CD ROS exerting a biological effect.
Collapse
Affiliation(s)
- Will Wheatley
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
31
|
Agostini S, Lionetti V. New insights into the non-hemostatic role of von Willebrand factor in endothelial protection. Can J Physiol Pharmacol 2017; 95:1183-1189. [PMID: 28715643 DOI: 10.1139/cjpp-2017-0126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During exposure to ischemia-reperfusion (I/R) insult, angiotensin II (AngII)-induced endothelin-1 (ET-1) upregulation in endothelial cells progressively impairs nitric oxide (NO) bioavailability while increasing levels of superoxide anion (O2-) and leading to the onset of endothelial dysfunction. Moreover, the overexpression of ET-1 increases the endothelial and circulating levels of von Willebrand factor (vWF), a glycoprotein with a crucial role in arterial thrombus formation. Nowadays, the non-hemostatic role of endothelial vWF is emerging, although we do not yet know whether its increased expression is cause or consequence of endothelial dysfunction. Notably, the vWF blockade or depletion leads to endothelial protection in cultured cells, animal models of vascular injury, and patients as well. Despite the recent efforts to develop an effective pharmacological strategy, the onset of endothelial dysfunction is still difficult to prevent and remains closely related to adverse clinical outcome. Unraveling the non-hemostatic role of endothelial vWF in the onset of endothelial dysfunction could provide new avenues for protection against vascular injury mediated by AngII.
Collapse
Affiliation(s)
- Silvia Agostini
- a Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Vincenzo Lionetti
- a Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,b UOS Anesthesiology, Fondazione Toscana "G. Monasterio", Pisa, Italy
| |
Collapse
|
32
|
Daiber A, Di Lisa F, Oelze M, Kröller‐Schön S, Steven S, Schulz E, Münzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol 2017; 174:1670-1689. [PMID: 26660451 PMCID: PMC5446573 DOI: 10.1111/bph.13403] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are associated with and/or caused by oxidative stress. This concept has been proven by using the approach of genetic deletion of reactive species producing (pro-oxidant) enzymes as well as by the overexpression of reactive species detoxifying (antioxidant) enzymes leading to a marked reduction of reactive oxygen and nitrogen species (RONS) and in parallel to an amelioration of the severity of diseases. Likewise, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of antioxidant RONS detoxifying enzymes. Thus, the consequences of the interaction (redox crosstalk) of superoxide/hydrogen peroxide produced by mitochondria with other ROS producing enzymes such as NADPH oxidases (Nox) are of outstanding importance and will be discussed including the consequences for endothelial nitric oxide synthase (eNOS) uncoupling as well as the redox regulation of the vascular function/tone in general (soluble guanylyl cyclase, endothelin-1, prostanoid synthesis). Pathways and potential mechanisms leading to this crosstalk will be analysed in detail and highlighted by selected examples from the current literature including hypoxia, angiotensin 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" will be discussed providing evidence that mitochondria represent key players and amplifiers of the burden of oxidative stress. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Fabio Di Lisa
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Matthias Oelze
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Swenja Kröller‐Schön
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Sebastian Steven
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Eberhard Schulz
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Thomas Münzel
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| |
Collapse
|
33
|
Daiber A, Steven S, Weber A, Shuvaev VV, Muzykantov VR, Laher I, Li H, Lamas S, Münzel T. Targeting vascular (endothelial) dysfunction. Br J Pharmacol 2017; 174:1591-1619. [PMID: 27187006 PMCID: PMC5446575 DOI: 10.1111/bph.13517] [Citation(s) in RCA: 352] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/28/2016] [Accepted: 05/09/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases are major contributors to global deaths and disability-adjusted life years, with hypertension a significant risk factor for all causes of death. The endothelium that lines the inner wall of the vasculature regulates essential haemostatic functions, such as vascular tone, circulation of blood cells, inflammation and platelet activity. Endothelial dysfunction is an early predictor of atherosclerosis and future cardiovascular events. We review the prognostic value of obtaining measurements of endothelial function, the clinical techniques for its determination, the mechanisms leading to endothelial dysfunction and the therapeutic treatment of endothelial dysfunction. Since vascular oxidative stress and inflammation are major determinants of endothelial function, we have also addressed current antioxidant and anti-inflammatory therapies. In the light of recent data that dispute the prognostic value of endothelial function in healthy human cohorts, we also discuss alternative diagnostic parameters such as vascular stiffness index and intima/media thickness ratio. We also suggest that assessing vascular function, including that of smooth muscle and even perivascular adipose tissue, may be an appropriate parameter for clinical investigations. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMainzGermany
| | - Sebastian Steven
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Alina Weber
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Vladimir V. Shuvaev
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Huige Li
- German Center for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMainzGermany
- Department of PharmacologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Santiago Lamas
- Department of Cell Biology and ImmunologyCentro de Biología Molecular "Severo Ochoa" (CSIC‐UAM)MadridSpain
| | - Thomas Münzel
- Center of CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Rhine‐MainMainzGermany
| |
Collapse
|
34
|
Pentaerythritol Tetranitrate In Vivo Treatment Improves Oxidative Stress and Vascular Dysfunction by Suppression of Endothelin-1 Signaling in Monocrotaline-Induced Pulmonary Hypertension. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4353462. [PMID: 28337251 PMCID: PMC5350298 DOI: 10.1155/2017/4353462] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/19/2017] [Indexed: 11/18/2022]
Abstract
Objective. Oxidative stress and endothelial dysfunction contribute to pulmonary arterial hypertension (PAH). The role of the nitrovasodilator pentaerythritol tetranitrate (PETN) on endothelial function and oxidative stress in PAH has not yet been defined. Methods and Results. PAH was induced by monocrotaline (MCT, i.v.) in Wistar rats. Low (30 mg/kg; MCT30), middle (40 mg/kg; MCT40), or high (60 mg/kg; MCT60) dose of MCT for 14, 28, and 42 d was used. MCT induced endothelial dysfunction, pulmonary vascular wall thickening, and fibrosis, as well as protein tyrosine nitration. Pulmonary arterial pressure and heart/body and lung/body weight ratio were increased in MCT40 rats (28 d) and reduced by oral PETN (10 mg/kg, 24 d) therapy. Oxidative stress in the vascular wall, in the heart, and in whole blood as well as vascular endothelin-1 signaling was increased in MCT40-treated rats and normalized by PETN therapy, likely by upregulation of heme oxygenase-1 (HO-1). PETN therapy improved endothelium-dependent relaxation in pulmonary arteries and inhibited endothelin-1-induced oxidative burst in whole blood and the expression of adhesion molecule (ICAM-1) in endothelial cells. Conclusion. MCT-induced PAH impairs endothelial function (aorta and pulmonary arteries) and increases oxidative stress whereas PETN markedly attenuates these adverse effects. Thus, PETN therapy improves pulmonary hypertension beyond its known cardiac preload reducing ability.
Collapse
|
35
|
The association of endothelin-1 with markers of oxidative stress in a biethnic South African cohort: the SABPA study. Hypertens Res 2016; 40:189-195. [DOI: 10.1038/hr.2016.128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/11/2016] [Accepted: 08/22/2016] [Indexed: 02/07/2023]
|
36
|
Gu Y, Wu J. Bovine lactoferrin-derived ACE inhibitory tripeptide LRP also shows antioxidative and anti-inflammatory activities in endothelial cells. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
37
|
Gene silencing of endothelial von Willebrand Factor attenuates angiotensin II-induced endothelin-1 expression in porcine aortic endothelial cells. Sci Rep 2016; 6:30048. [PMID: 27443965 PMCID: PMC4957110 DOI: 10.1038/srep30048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022] Open
Abstract
Expression of endothelin (ET)-1 is increased in endothelial cells exposed to angiotensin II (Ang II), leading to endothelial dysfunction and cardiovascular disorders. Since von Willebrand Factor (vWF) blockade improves endothelial function in coronary patients, we hypothesized that targeting endothelial vWF with short interference RNA (siRNA) prevents Ang II-induced ET-1 upregulation. Nearly 65 ± 2% silencing of vWF in porcine aortic endothelial cells (PAOECs) was achieved with vWF-specific siRNA without affecting cell viability and growth. While showing ET-1 similar to wild type cells at rest, vWF-silenced cells did not present ET-1 upregulation during exposure to Ang II (100 nM/24 h), preserving levels of endothelial nitric oxide synthase activity similar to wild type. vWF silencing prevented AngII-induced increase in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity and superoxide anion (O2-) levels, known triggers of ET-1 expression. Moreover, no increase in O2- or ET-1 levels was found in silenced cells treated with AngII or NOX-agonist phorbol ester (PMA 5 nM/48 h). Finally, vWF was required for overexpression of NOX4 and NOX2 in response to AngII and PMA. In conclusion, endothelial vWF knockdown prevented Ang II-induced ET-1 upregulation through attenuation of NOX-mediated O2- production. Our findings reveal a new role of vWF in preventing of Ang II-induced endothelial dysfunction.
Collapse
|
38
|
Hobson SR, Acharya R, Lim R, Chan ST, Mockler J, Wallace EM. Role of activin A in the pathogenesis of endothelial cell dysfunction in preeclampsia. Pregnancy Hypertens 2016; 6:130-3. [DOI: 10.1016/j.preghy.2016.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/10/2016] [Accepted: 03/29/2016] [Indexed: 11/26/2022]
|
39
|
Endothelin-1 Expression Associated with Lipid Peroxidation and Nuclear Factor-κB Activation in Type 2 Diabetes Mellitus Patients with Angiopathy and Limb Amputation. Plast Reconstr Surg 2016; 137:187e-195e. [DOI: 10.1097/prs.0000000000001886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
40
|
Daiber A, Münzel T. Organic Nitrate Therapy, Nitrate Tolerance, and Nitrate-Induced Endothelial Dysfunction: Emphasis on Redox Biology and Oxidative Stress. Antioxid Redox Signal 2015; 23:899-942. [PMID: 26261901 PMCID: PMC4752190 DOI: 10.1089/ars.2015.6376] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Organic nitrates, such as nitroglycerin (GTN), isosorbide-5-mononitrate and isosorbide dinitrate, and pentaerithrityl tetranitrate (PETN), when given acutely, have potent vasodilator effects improving symptoms in patients with acute and chronic congestive heart failure, stable coronary artery disease, acute coronary syndromes, or arterial hypertension. The mechanisms underlying vasodilation include the release of •NO or a related compound in response to intracellular bioactivation (for GTN, the mitochondrial aldehyde dehydrogenase [ALDH-2]) and activation of the enzyme, soluble guanylyl cyclase. Increasing cyclic guanosine-3',-5'-monophosphate (cGMP) levels lead to an activation of the cGMP-dependent kinase I, thereby causing the relaxation of the vascular smooth muscle by decreasing intracellular calcium concentrations. The hemodynamic and anti-ischemic effects of organic nitrates are rapidly lost upon long-term (low-dose) administration due to the rapid development of tolerance and endothelial dysfunction, which is in most cases linked to increased intracellular oxidative stress. Enzymatic sources of reactive oxygen species under nitrate therapy include mitochondria, NADPH oxidases, and an uncoupled •NO synthase. Acute high-dose challenges with organic nitrates cause a similar loss of potency (tachyphylaxis), but with distinct pathomechanism. The differences among organic nitrates are highlighted regarding their potency to induce oxidative stress and subsequent tolerance and endothelial dysfunction. We also address pleiotropic effects of organic nitrates, for example, their capacity to stimulate antioxidant pathways like those demonstrated for PETN, all of which may prevent adverse effects in response to long-term therapy. Based on these considerations, we will discuss and present some preclinical data on how the nitrate of the future should be designed.
Collapse
Affiliation(s)
- Andreas Daiber
- The 2nd Medical Clinic, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| | - Thomas Münzel
- The 2nd Medical Clinic, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| |
Collapse
|
41
|
Wierzbowska J, Wojtkiewicz S, Zbieć A, Wierzbowski R, Liebert A, Maniewski R. Prolonged postocclusive hyperemia response in patients with normal-tension glaucoma. Med Sci Monit 2014; 20:2607-16. [PMID: 25502623 PMCID: PMC4266367 DOI: 10.12659/msm.891069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background It is believed that endothelial dysfunction may be a link between systemic and ocular dysregulation in glaucoma. The aim of this study was to evaluate peripheral vascular reactive hyperemia in response to occlusion test and to correlate peripheral vascular findings with retrobulbar hemodynamics parameters in patients with normal-tension glaucoma. Material/Methods Forty-eight patients with normal-tension glaucoma (mean age 58.1 years, 38 women) and 40 control subjects (mean age 54.1 years, 36 women) were subjected to a brachial arterial occlusion test and color Doppler imaging (LOGIQ 9, GE Medical Systems) of the retrobulbar arteries. Finger hyperemia was assessed by using a 2-channel laser Doppler flowmeter (MBF-3D, Moor Instruments, Ltd.). Time parameters (time to peak flow, half-time of hyperemia, time of recovery) and amplitude parameters (maximum hyperemia response, biological zero) of the post-occlusive reactive hyperemia signal pattern as well as velocities and resistance index of the ophthalmic, central retinal, and short posterior ciliary arteries were evaluated and compared between study groups. Results In glaucoma patients, time to peak flow and half-time of hyperemia were significantly longer (21.4 vs. 12.0 s, p=0.02 and 74.1 vs. 44.2 s, p=0.03, respectively) and biological zero was significantly lower (2.4 vs. 3.2, p=0.01) comparing with healthy subjects. In glaucoma patients, peak-systolic and end-diastolic velocities of central retinal artery were significantly lower (12.8 vs.14.1, p=0.03 and 3.9 vs. 4.7, p=0.01, respectively) and resistance index of this artery was significantly higher (0.69 vs. 0.67, p=0.03) compared to controls. In the glaucoma group, maximum hyperemic response was negatively correlated with the resistance index of temporal short posterior ciliary arteries (r=−0.4, p=0.01), whereas in the control group half-time of hyperemia was negatively correlated with end-diastolic velocity of the central retinal artery (r=−0.3, p=0.03). Conclusions Arterial occlusion test elicited a prolonged systemic hyperemia response in patients with glaucoma as compared with healthy subjects. Retrobulbar blood flow alterations in glaucoma patients may be related to systemic vascular dysregulation.
Collapse
Affiliation(s)
- Joanna Wierzbowska
- Department of Ophthalmology, Military Institute of Medicine, Warsaw, Poland
| | - Stanisław Wojtkiewicz
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Zbieć
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Robert Wierzbowski
- Department of Cardiology, Military Institute of Medicine, Warsaw, Poland
| | - Adam Liebert
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Roman Maniewski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
42
|
Münzel T, Steven S, Daiber A. Organic nitrates: update on mechanisms underlying vasodilation, tolerance and endothelial dysfunction. Vascul Pharmacol 2014; 63:105-13. [PMID: 25446162 DOI: 10.1016/j.vph.2014.09.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/15/2014] [Accepted: 09/20/2014] [Indexed: 10/24/2022]
Abstract
Given acutely, organic nitrates, such as nitroglycerin (GTN), isosorbide mono- and dinitrates (ISMN, ISDN), and pentaerythrityl tetranitrate (PETN), have potent vasodilator and anti-ischemic effects in patients with acute coronary syndromes, acute and chronic congestive heart failure and arterial hypertension. During long-term treatment, however, side effects such as nitrate tolerance and endothelial dysfunction occur, and therapeutic efficacy of these drugs rapidly vanishes. Recent experimental and clinical studies have revealed that organic nitrates per se are not just nitric oxide (NO) donors, but rather a quite heterogeneous group of drugs considerably differing for mechanisms underlying vasodilation and the development of endothelial dysfunction and tolerance. Based on this, we propose that the term nitrate tolerance should be avoided and more specifically the terms of GTN, ISMN and ISDN tolerance should be used. The present review summarizes preclinical and clinical data concerning organic nitrates. Here we also emphasize the consequences of chronic nitrate therapy on the supersensitivity of the vasculature to vasoconstriction and on the increased autocrine expression of endothelin. We believe that these so far rather neglected and underestimated side effects of chronic therapy with at least GTN and ISMN are clinically important.
Collapse
Affiliation(s)
- Thomas Münzel
- Department of Cardiology and Angiology, University Medical Center, Mainz, Germany.
| | - Sebastian Steven
- Department of Cardiology and Angiology, University Medical Center, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology and Angiology, University Medical Center, Mainz, Germany
| |
Collapse
|
43
|
Matsumoto T, Lopes RAM, Taguchi K, Kobayashi T, Tostes RC. Linking the beneficial effects of current therapeutic approaches in diabetes to the vascular endothelin system. Life Sci 2014; 118:129-35. [PMID: 24418002 DOI: 10.1016/j.lfs.2013.12.216] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/04/2013] [Accepted: 12/24/2013] [Indexed: 12/19/2022]
Abstract
The rising epidemic of diabetes worldwide is of significant concern. Although the ultimate objective is to prevent the development and find a cure for the disease, prevention and treatment of diabetic complications is very important. Vascular complications in diabetes, or diabetic vasculopathy, include macro- and microvascular dysfunction and represent the principal cause of morbidity and mortality in diabetic patients. Endothelial dysfunction plays a pivotal role in the development and progression of diabetic vasculopathy. Endothelin-1 (ET-1), an endothelial cell-derived peptide, is a potent vasoconstrictor with mitogenic, pro-oxidative and pro-inflammatory properties that are particularly relevant to the pathophysiology of diabetic vasculopathy. Overproduction of ET-1 is reported in patients and animal models of diabetes and the functional effects of ET-1 and its receptors are also greatly altered in diabetic conditions. The current therapeutic approaches in diabetes include glucose lowering, sensitization to insulin, reduction of fatty acids and vasculoprotective therapies. However, whether and how these therapeutic approaches affect the ET-1 system remain poorly understood. Accordingly, in the present review, we will focus on experimental and clinical evidence that indicates a role for ET-1 in diabetic vasculopathy and on the effects of current therapeutic approaches in diabetes on the vascular ET-1 system.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Rheure A M Lopes
- Department of Pharmacology, Medical School of Ribeirao Preto, University of Sao Paulo, Av Bandeirantes 3900, Ribeirao Preto, SP 14049-900, Brazil
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Rita C Tostes
- Department of Pharmacology, Medical School of Ribeirao Preto, University of Sao Paulo, Av Bandeirantes 3900, Ribeirao Preto, SP 14049-900, Brazil
| |
Collapse
|
44
|
Wang HD, Chapman A. Essential Role of Adventitial Reactive Oxygen Species (ROS) in Vascular Function. SYSTEMS BIOLOGY OF FREE RADICALS AND ANTIOXIDANTS 2014:1287-1303. [DOI: 10.1007/978-3-642-30018-9_54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
45
|
Friedman JK, Nitta CH, Henderson KM, Codianni SJ, Sanchez L, Ramiro-Diaz JM, Howard TA, Giermakowska W, Kanagy NL, Gonzalez Bosc LV. Intermittent hypoxia-induced increases in reactive oxygen species activate NFATc3 increasing endothelin-1 vasoconstrictor reactivity. Vascul Pharmacol 2013; 60:17-24. [PMID: 24239798 DOI: 10.1016/j.vph.2013.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 10/14/2013] [Accepted: 11/05/2013] [Indexed: 02/09/2023]
Abstract
Sleep apnea (SA), defined as intermittent respiratory arrest during sleep, is associated with increased incidence of hypertension, peripheral vascular disease, stroke, and sudden cardiac death. We have shown that intermittent hypoxia with CO2 supplementation (IH), a model for SA, increases blood pressure and circulating ET-1 levels, upregulates lung pre-pro ET-1 mRNA, increases vasoconstrictor reactivity to ET-1 in rat small mesenteric arteries (MA) and increases vascular reactive oxygen species (ROS). NFAT activity is increased in the aorta (AO) and MA of mice exposed to IH in an ET-1-dependent manner, and the genetic ablation of the isoform NFATc3 prevents IH-induced hypertension. We hypothesized that IH causes an increase in arterial ROS generation, which activates NFATc3 to increase vasoconstrictor reactivity to ET-1. In support of our hypothesis, we found that IH increases ROS in AO and MA. In vivo administration of the SOD mimetic tempol during IH exposure prevents IH-induced increases in NFAT activity in mouse MA and AO. We found that IH causes an NFATc3-dependent increase in vasoconstrictor reactivity to ET-1, accompanied by an increase in vessel wall [Ca²⁺]. Our results indicate that IH exposure causes an increase in arterial ROS to activate NFATc3, which then increases vasoconstrictor reactivity and Ca²⁺ response to ET-1. These studies highlight a novel regulatory pathway, and demonstrate the potential clinical relevance of NFAT inhibition to prevent hypertension in SA patients.
Collapse
Affiliation(s)
- J K Friedman
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - C H Nitta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - K M Henderson
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - S J Codianni
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - L Sanchez
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - J M Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - T A Howard
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - W Giermakowska
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - N L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - L V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
46
|
Schnabel RB, Wild PS, Schulz A, Zeller T, Sinning CR, Wilde S, Kunde J, Lubos E, Lackner KJ, Warnholtz A, Gori T, Blankenberg S, Munzel T. Multiple endothelial biomarkers and noninvasive vascular function in the general population: the Gutenberg Health Study. Hypertension 2012; 60:288-95. [PMID: 22689741 DOI: 10.1161/hypertensionaha.112.191874] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vascular reactivity is reflected by blood biomarkers and noninvasive vascular function measurement. The relation of biomarkers to flow-mediated dilation and peripheral arterial tonometry in the general population is little understood. In 5000 individuals (mean age, 56±11 years; age range, 35-74 years; 49% women) of the population-based Gutenberg Health Study we simultaneously assessed 6 biomarkers of cardiovascular function (midregional proadrenomedullin [MR-proADM], midregional pro atrial natriuretic peptide [MR-proANP], N-terminal pro B-type natriuretic peptide, copeptin, C-terminal proendothelin 1, and neopterin) in relation to flow-mediated dilation and peripheral arterial tonometry. Strongest partial correlations (adjusted for age and sex) were observed for baseline pulse amplitude with MR-proADM (r=0.13) and MR-proANP (r=-0.13); hyperemic response variables showed the highest correlation for MR-proADM and peripheral arterial tonometry ratio (r=-0.14). In multivariable linear regression models, strongest associations with baseline vascular function were observed for MR-proANP with baseline pulse amplitude (β per SD increase [99.17%], -0.080 [-0.115 to -0.044]; P<0.0001 after Bonferroni correction for multiple testing) and MR-proADM (-0.044 [-0.070 to -0.017]; P<0.0001), as well as MR-proANP (-0.033 [-0.057 to -0.009]; P=0.0017) and N-terminal pro B-type natriuretic peptide (-0.027 [-0.051 to -0.003]; P=0.015) with brachial artery diameter. For hyperemic response variables, highest associations were seen for peripheral arterial tonometry ratio with MR-proADM (-0.022 [-0.043 to -0.004]; P=0.043), MR-proANP (0.016 [-0.0034 to 0.035]; P=0.18), and C-terminal proendothelin 1 (-0.025 [-0.043 to -0.008]; P=0.00094]. In our large, population-based study, we identified MR-proADM and MR-proANP as circulating biomarkers of vascular function most strongly related to noninvasive measures of conduit artery and peripheral arterial performance. Whether determination of blood biomarkers helps to better understand vascular pathology and may provide prognostic information needs to be investigated in future studies.
Collapse
Affiliation(s)
- Renate B Schnabel
- University Heart Center, Department of General and Interventional Cardiology, Martinistr 52, 20246 Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Oelze M, Knorr M, Kröller-Schön S, Kossmann S, Gottschlich A, Rümmler R, Schuff A, Daub S, Doppler C, Kleinert H, Gori T, Daiber A, Münzel T. Chronic therapy with isosorbide-5-mononitrate causes endothelial dysfunction, oxidative stress, and a marked increase in vascular endothelin-1 expression. Eur Heart J 2012; 34:3206-16. [PMID: 22555214 DOI: 10.1093/eurheartj/ehs100] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Isosorbide-5-mononitrate (ISMN) is one of the most frequently used compounds in the treatment of coronary artery disease predominantly in the USA. However, ISMN was reported to induce endothelial dysfunction, which was corrected by vitamin C pointing to a crucial role of reactive oxygen species (ROS) in causing this phenomenon. We sought to elucidate the mechanism how ISMN causes endothelial dysfunction and oxidative stress in vascular tissue. METHODS AND RESULTS Male Wistar rats (n= 69 in total) were treated with ISMN (75 mg/kg/day) or placebo for 7 days. Endothelin (ET) expression was determined by immunohistochemistry in aortic sections. Isosorbide-5-mononitrate infusion caused significant endothelial dysfunction but no tolerance to ISMN itself, whereas ROS formation and nicotinamide adenine dinucleotidephosphate (NADPH) oxidase activity in the aorta, heart, and whole blood were increased. Isosorbide-5-mononitrate up-regulated the expression of NADPH subunits and caused uncoupling of the endothelial nitric oxide synthase (eNOS) likely due to a down-regulation of the tetrahydrobiopterin-synthesizing enzyme GTP-cyclohydrolase-1 and to S-glutathionylation of eNOS. The adverse effects of ISMN were improved in gp91phox knockout mice and normalized by bosentan in vivo/ex vivo treatment and suppressed by apocynin. In addition, a strong increase in the expression of ET within the endothelial cell layer and the adventitia was observed. CONCLUSION Chronic treatment with ISMN causes endothelial dysfunction and oxidative stress, predominantly by an ET-dependent activation of the vascular and phagocytic NADPH oxidase activity and NOS uncoupling. These findings may explain at least in part results from a retrospective analysis indicating increased mortality in post-infarct patients in response to long-term treatment with mononitrates.
Collapse
Affiliation(s)
- Matthias Oelze
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Cho SC, Rhim JH, Choi HR, Son YH, Lee SJ, Song KY, Park SC. Protective effect of 4,4'-diaminodiphenylsulfone against paraquat-induced mouse lung injury. Exp Mol Med 2012; 43:525-37. [PMID: 21765237 DOI: 10.3858/emm.2011.43.9.060] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Although 4,4'-diaminodiphenylsulfone (DDS, dapsone) has been used to treat several dermatologic conditions, including Hansen disease, for the past several decades, its mode of action has remained a topic of debate. We recently reported that DDS treatment significantly extends the lifespan of the nematode C. elegans by decreasing the generation of reactive oxygen species. Additionally, in in vitro experiments using non-phagocytic human fibroblasts, we found that DDS effectively counteracted the toxicity of paraquat (PQ). In the present study, we extended our work to test the protective effect of DDS against PQ in vivo using a mouse lung injury model. Oral administration of DDS to mice significantly attenuated the lung tissue damage caused by subsequent administration of PQ. Moreover, DDS reduced the local expression of mRNA transcripts encoding inflammation-related molecules, including endothelin-1 (ET-1), macrophage inflammatory protein-1α (MIP-1α), and transforming growth factor-β (TGF-β). In addition, DDS decreased the PQ-induced expression of NADPH oxidase mRNA and activation of protein kinase Cμ (PKCμ). DDS treatment also decreased the PQ-induced generation of superoxide anions in mouse lung fibroblasts. Taken together, these data suggest the novel efficacy of DDS as an effective protective agent against oxidative stress-induced tissue damages.
Collapse
Affiliation(s)
- Sung Chun Cho
- Department of Biochemistry and Molecular Biology, Institute on Aging, Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
The discovery of endothelium-dependent contraction: The legacy of Paul M. Vanhoutte. Pharmacol Res 2011; 63:455-62. [DOI: 10.1016/j.phrs.2011.02.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Accepted: 02/28/2011] [Indexed: 01/10/2023]
|
50
|
|