1
|
Power G, Ferreira-Santos L, Martinez-Lemus LA, Padilla J. Integrating molecular and cellular components of endothelial shear stress mechanotransduction. Am J Physiol Heart Circ Physiol 2024; 327:H989-H1003. [PMID: 39178024 PMCID: PMC11482243 DOI: 10.1152/ajpheart.00431.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
The lining of blood vessels is constantly exposed to mechanical forces exerted by blood flow against the endothelium. Endothelial cells detect these tangential forces (i.e., shear stress), initiating a host of intracellular signaling cascades that regulate vascular physiology. Thus, vascular health is tethered to the endothelial cells' capacity to transduce shear stress. Indeed, the mechanotransduction of shear stress underlies a variety of cardiovascular benefits, including some of those associated with increased physical activity. However, endothelial mechanotransduction is impaired in aging and disease states such as obesity and type 2 diabetes, precipitating the development of vascular disease. Understanding endothelial mechanotransduction of shear stress, and the molecular and cellular mechanisms by which this process becomes defective, is critical for the identification and development of novel therapeutic targets against cardiovascular disease. In this review, we detail the primary mechanosensitive structures that have been implicated in detecting shear stress, including junctional proteins such as platelet endothelial cell adhesion molecule-1 (PECAM-1), the extracellular glycocalyx and its components, and ion channels such as piezo1. We delineate which molecules are truly mechanosensitive and which may simply be indispensable for the downstream transmission of force. Furthermore, we discuss how these mechanosensors interact with other cellular structures, such as the cytoskeleton and membrane lipid rafts, which are implicated in translating shear forces to biochemical signals. Based on findings to date, we also seek to integrate these cellular and molecular mechanisms with a view of deciphering endothelial mechanotransduction of shear stress, a tenet of vascular physiology.
Collapse
Affiliation(s)
- Gavin Power
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | | | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| |
Collapse
|
2
|
Martin E, Girardello R, Dittmar G, Ludwig A. Time-resolved proximity proteomics uncovers a membrane tension-sensitive caveolin-1 interactome at the rear of migrating cells. eLife 2024; 13:e85601. [PMID: 39315773 PMCID: PMC11509677 DOI: 10.7554/elife.85601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/23/2024] [Indexed: 09/25/2024] Open
Abstract
Caveolae are small membrane pits with fundamental roles in mechanotransduction. Several studies have shown that caveolae flatten out in response to increased membrane tension, thereby acting as a mechanosensitive membrane reservoir that buffers acute mechanical stress. Caveolae have also been implicated in the control of RhoA/ROCK-mediated actomyosin contractility at the rear of migrating cells. However, how membrane tension controls the organisation of caveolae and their role in mechanotransduction remains unclear. To address this, we systematically quantified protein-protein interactions of caveolin-1 in migrating RPE1 cells at steady state and in response to an acute increase in membrane tension using biotin-based proximity labelling and quantitative mass spectrometry. Our data show that caveolae are highly enriched at the rear of migrating RPE1 cells and that membrane tension rapidly and reversibly disrupts the caveolar protein coat. Membrane tension also detaches caveolin-1 from focal adhesion proteins and several mechanosensitive regulators of cortical actin including filamins and cortactin. In addition, we present evidence that ROCK and the RhoGAP ARHGAP29 associate with caveolin-1 in a manner dependent on membrane tension, with ARHGAP29 influencing caveolin-1 Y14 phosphorylation, caveolae rear localisation, and RPE1 cell migration. Taken together, our work uncovers a membrane tension-sensitive coupling between caveolae and the rear-localised F-actin cytoskeleton. This provides a framework for dissecting the molecular mechanisms underlying caveolae-regulated mechanotransduction pathways.
Collapse
Affiliation(s)
- Eleanor Martin
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- NTU Institute of Structural Biology (NISB), Nanyang Technological University, Singapore, Singapore
| | - Rossana Girardello
- Proteomics of Cellular Signaling, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Gunnar Dittmar
- Proteomics of Cellular Signaling, Luxembourg Institute of Health, Strassen, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- NTU Institute of Structural Biology (NISB), Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
3
|
Hong SG, Ashby JW, Kennelly JP, Wu M, Steel M, Chattopadhyay E, Foreman R, Tontonoz P, Tarling EJ, Turowski P, Gallagher-Jones M, Mack JJ. Mechanosensitive membrane domains regulate calcium entry in arterial endothelial cells to protect against inflammation. J Clin Invest 2024; 134:e175057. [PMID: 38771648 PMCID: PMC11213468 DOI: 10.1172/jci175057] [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: 08/23/2023] [Accepted: 05/10/2024] [Indexed: 05/23/2024] Open
Abstract
Endothelial cells (ECs) in the descending aorta are exposed to high laminar shear stress, and this supports an antiinflammatory phenotype. High laminar shear stress also induces flow-aligned cell elongation and front-rear polarity, but whether these are required for the antiinflammatory phenotype is unclear. Here, we showed that caveolin-1-rich microdomains polarize to the downstream end of ECs that are exposed to continuous high laminar flow. These microdomains were characterized by high membrane rigidity, filamentous actin (F-actin), and raft-associated lipids. Transient receptor potential vanilloid (TRPV4) ion channels were ubiquitously expressed on the plasma membrane but mediated localized Ca2+ entry only at these microdomains where they physically interacted with clustered caveolin-1. These focal Ca2+ bursts activated endothelial nitric oxide synthase within the confines of these domains. Importantly, we found that signaling at these domains required both cell body elongation and sustained flow. Finally, TRPV4 signaling at these domains was necessary and sufficient to suppress inflammatory gene expression and exogenous activation of TRPV4 channels ameliorated the inflammatory response to stimuli both in vitro and in vivo. Our work revealed a polarized mechanosensitive signaling hub in arterial ECs that dampened inflammatory gene expression and promoted cell resilience.
Collapse
Affiliation(s)
- Soon-Gook Hong
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| | | | - John P. Kennelly
- Molecular Biology Institute
- Department of Pathology and Laboratory Medicine, and
| | - Meigan Wu
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| | | | | | - Rob Foreman
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, California, USA
| | - Peter Tontonoz
- Molecular Biology Institute
- Department of Pathology and Laboratory Medicine, and
| | | | - Patric Turowski
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Marcus Gallagher-Jones
- Correlated Imaging, Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, United Kingdom
| | - Julia J. Mack
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| |
Collapse
|
4
|
Mirza I, Haloul M, Hassan C, Masrur M, Mostafa A, Bianco FM, Ali MM, Minshall RD, Mahmoud AM. Adiposomes from Obese-Diabetic Individuals Promote Endothelial Dysfunction and Loss of Surface Caveolae. Cells 2023; 12:2453. [PMID: 37887297 PMCID: PMC10605845 DOI: 10.3390/cells12202453] [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: 09/22/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Glycosphingolipids (GSLs) are products of lipid glycosylation that have been implicated in the development of cardiovascular diseases. In diabetes, the adipocyte microenvironment is characterized by hyperglycemia and inflammation, resulting in high levels of GSLs. Therefore, we sought to assess the GSL content in extracellular vesicles derived from the adipose tissues (adiposomes) of obese-diabetic (OB-T2D) subjects and their impact on endothelial cell function. To this end, endothelial cells were exposed to adiposomes isolated from OB-T2D versus healthy subjects. Cells were assessed for caveolar integrity and related signaling, such as Src-kinase and caveolin-1 (cav-1) phosphorylation, and functional pathways, such as endothelial nitric oxide synthase (eNOS) activity. Compared with adiposomes from healthy subjects, OB-T2D adiposomes had higher levels of GSLs, especially LacCer and GM3; they promoted cav-1 phosphorylation coupled to an obvious loss of endothelial surface caveolae and induced eNOS-uncoupling, peroxynitrite generation, and cav-1 nitrosylation. These effects were abolished by Src kinase inhibition and were not observed in GSL-depleted adiposomes. At the functional levels, OB-T2D adiposomes reduced nitric oxide production, shear response, and albumin intake in endothelial cells and impaired flow-induced dilation in healthy arterioles. In conclusion, OB-T2D adiposomes carried a detrimental GSL cargo that disturbed endothelial caveolae and the associated signaling.
Collapse
Affiliation(s)
- Imaduddin Mirza
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (I.M.); (M.H.)
| | - Mohamed Haloul
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (I.M.); (M.H.)
| | - Chandra Hassan
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Mario Masrur
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Amro Mostafa
- Departments of Anesthesiology and Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco M. Bianco
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Mohamed M. Ali
- School of Business and Non-Profit Management, North Park University, Chicago, IL 60625, USA;
| | - Richard D. Minshall
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Abeer M. Mahmoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (I.M.); (M.H.)
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Shu Y, Jin S. Caveolin-1 in endothelial cells: A potential therapeutic target for atherosclerosis. Heliyon 2023; 9:e18653. [PMID: 37554846 PMCID: PMC10405014 DOI: 10.1016/j.heliyon.2023.e18653] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/10/2023] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease characterized by lipid accumulation and the activation of the inflammatory response; it remains the leading nation-wide cause of death. Early in the progression of AS, stimulation by pro-inflammatory agonists (TNF-α, LPS, and others), oxidized lipoproteins (ox-LDL), and biomechanical stimuli (low shear stress) lead to endothelial cell activation and dysfunction. Consequently, it is crucial to investigate how endothelial cells respond to different stressors and ways to alter endothelial cell activation in AS development, as they are the earliest cells to respond. Caveolin-1 (Cav1) is a 21-24-kDa membrane protein located in caveolae and highly expressed in endothelial cells, which plays a vital role in regulating lipid transport, inflammatory responses, and various cellular signaling pathways and has atherogenic effects. This review summarizes recent studies on the structure and physiological functions of Cav1 and outlines the potential mechanisms it mediates in AS development. Included are the roles of Cav1 in the regulation of endothelial cell autophagy, response to shear stress, modulation of the eNOS/NO axis, and transduction of inflammatory signaling pathways. This review provides a rationale for proposing Cav1 as a novel target for the prevention of AS, as well as new ideas for therapeutic strategies for early AS.
Collapse
Affiliation(s)
- Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, 430077, China
| |
Collapse
|
6
|
Aitken C, Mehta V, Schwartz MA, Tzima E. Mechanisms of endothelial flow sensing. NATURE CARDIOVASCULAR RESEARCH 2023; 2:517-529. [PMID: 39195881 DOI: 10.1038/s44161-023-00276-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/14/2023] [Indexed: 08/29/2024]
Abstract
Fluid shear stress plays a key role in sculpting blood vessels during development, in adult vascular homeostasis and in vascular pathologies. During evolution, endothelial cells evolved several mechanosensors that convert physical forces into biochemical signals, a process termed mechanotransduction. This Review discusses our understanding of endothelial flow sensing and suggests important questions for future investigation.
Collapse
Affiliation(s)
- Claire Aitken
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vedanta Mehta
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - Ellie Tzima
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Hong SG, Ashby JW, Kennelly JP, Wu M, Chattopadhyay E, Foreman R, Tontonoz P, Turowski P, Gallagher-Jones M, Mack JJ. Polarized Mechanosensitive Signaling Domains Protect Arterial Endothelial Cells Against Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542500. [PMID: 37292837 PMCID: PMC10246006 DOI: 10.1101/2023.05.26.542500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Endothelial cells (ECs) in the descending aorta are exposed to high laminar shear stress, which supports an anti-inflammatory phenotype that protects them from atherosclerosis. High laminar shear stress also supports flow-aligned cell elongation and front-rear polarity, but whether this is required for athero-protective signaling is unclear. Here, we show that Caveolin-1-rich microdomains become polarized at the downstream end of ECs exposed to continuous high laminar flow. These microdomains are characterized by higher membrane rigidity, filamentous actin (F-actin) and lipid accumulation. Transient receptor potential vanilloid-type 4 (Trpv4) ion channels, while ubiquitously expressed, mediate localized Ca 2+ entry at these microdomains where they physically interact with clustered Caveolin-1. The resultant focal bursts in Ca 2+ activate the anti-inflammatory factor endothelial nitric oxide synthase (eNOS) within the confines of these domains. Importantly, we find that signaling at these domains requires both cell body elongation and sustained flow. Finally, Trpv4 signaling at these domains is necessary and sufficient to suppress inflammatory gene expression. Our work reveals a novel polarized mechanosensitive signaling hub that induces an anti-inflammatory response in arterial ECs exposed to high laminar shear stress.
Collapse
|
8
|
Effects of shear stress on vascular endothelial functions in atherosclerosis and potential therapeutic approaches. Biomed Pharmacother 2023; 158:114198. [PMID: 36916427 DOI: 10.1016/j.biopha.2022.114198] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/09/2022] [Accepted: 12/29/2022] [Indexed: 01/07/2023] Open
Abstract
Different blood flow patterns in the arteries can alter the adaptive phenotype of vascular endothelial cells (ECs), thereby affecting the functions of ECs and are directly associated with the occurrence of lesions in the early stages of atherosclerosis (AS). Atherosclerotic plaques are commonly found at curved or bifurcated arteries, where the blood flow pattern is dominated by oscillating shear stress (OSS). OSS can induce ECs to transform into pro-inflammatory phenotypes, increase cellular inflammation, oxidative stress response, mitochondrial dysfunction, metabolic abnormalities and endothelial permeability, thereby promoting the progression of AS. On the other hand, the straight artery has a stable laminar shear stress (LSS), which promotes the transformation of ECs into an anti-inflammatory phenotype, improves endothelial cell function, thereby inhibits atherosclerotic progression. ECs have the ability to actively sense, integrate, and convert mechanical stimuli by shear stress into biochemical signals that further induces intracellular changes (such as the opening and closing of ion channels, activation and transcription of signaling pathways). Here we not only outline the relationship between functions of vascular ECs and different forms of fluid shear stress in AS, but also aim to provide new solutions for potential atherosclerotic therapies targeting intracellular mechanical transductions.
Collapse
|
9
|
Enyong EN, Gurley JM, De Ieso ML, Stamer WD, Elliott MH. Caveolar and non-Caveolar Caveolin-1 in ocular homeostasis and disease. Prog Retin Eye Res 2022; 91:101094. [PMID: 35729002 PMCID: PMC9669151 DOI: 10.1016/j.preteyeres.2022.101094] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
Caveolae, specialized plasma membrane invaginations present in most cell types, play important roles in multiple cellular processes including cell signaling, lipid uptake and metabolism, endocytosis and mechanotransduction. They are found in almost all cell types but most abundant in endothelial cells, adipocytes and fibroblasts. Caveolin-1 (Cav1), the signature structural protein of caveolae was the first protein associated with caveolae, and in association with Cavin1/PTRF is required for caveolae formation. Genetic ablation of either Cav1 or Cavin1/PTRF downregulates expression of the other resulting in loss of caveolae. Studies using Cav1-deficient mouse models have implicated caveolae with human diseases such as cardiomyopathies, lipodystrophies, diabetes and muscular dystrophies. While caveolins and caveolae are extensively studied in extra-ocular settings, their contributions to ocular function and disease pathogenesis are just beginning to be appreciated. Several putative caveolin/caveolae functions are relevant to the eye and Cav1 is highly expressed in retinal vascular and choroidal endothelium, Müller glia, the retinal pigment epithelium (RPE), and the Schlemm's canal endothelium and trabecular meshwork cells. Variants at the CAV1/2 gene locus are associated with risk of primary open angle glaucoma and the high risk HTRA1 variant for age-related macular degeneration is thought to exert its effect through regulation of Cav1 expression. Caveolins also play important roles in modulating retinal neuroinflammation and blood retinal barrier permeability. In this article, we describe the current state of caveolin/caveolae research in the context of ocular function and pathophysiology. Finally, we discuss new evidence showing that retinal Cav1 exists and functions outside caveolae.
Collapse
Affiliation(s)
- Eric N Enyong
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jami M Gurley
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael L De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, USA
| | - Michael H Elliott
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
10
|
Liu W, Song H, Xu J, Guo Y, Zhang C, Yao Y, Zhang H, Liu Z, Li YC. Low shear stress inhibits endothelial mitophagy via caveolin-1/miR-7-5p/SQSTM1 signaling pathway. Atherosclerosis 2022; 356:9-17. [PMID: 35952464 DOI: 10.1016/j.atherosclerosis.2022.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 06/20/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Mitophagy plays a crucial role in mitochondrial homeostasis and is closely associated with endothelial function. However, the mechanism underlying low blood flow shear stress (SS), detrimental cellular stress, regulating endothelial mitophagy is unclear. This study aimed to investigate whether low SS inhibits endothelial mitophagy via caveolin-1 (Cav-1)/miR-7-5p/Sequestosome 1 (SQSTM1) signaling pathway. METHODS Low SS in vivo modeling was induced using a perivascular SS modifier implanted in the carotid artery of mice. In vitro modeling, low and physiological SS (4 and 15 dyn/cm2, respectively) were exerted on human aortic endothelial cells using a parallel plate chamber system. RESULTS Compared with physiological SS, low SS significantly inhibited endothelial mitophagy shown by down-regulation of SQSTM1, PINK1, Parkin, and LC 3II expressions. Deficient mitophagy deteriorated mitochondrial dynamics shown by up-regulation of Mfn1 and Fis1 expression and led to decreases in mitochondrial membrane potential. Cav-1 plays a bridging role in the process of low SS inhibiting mitophagy. The up-regulated miR-7-5p expression induced by low SS was suppressed after Cav-1 was silenced. The results of dual-luciferase reporter assays showed that miR-7-5p targeted inhibiting the SQSTM1 gene. Oxidative stress reaction shown by the elevation of reactive oxygen species and O2●- and endothelial dysfunction by the decrease in nitric oxide and the increase in macrophage chemoattractant protein-1 were associated with the low SS inhibiting endothelial mitophagy process. CONCLUSIONS Cav-1/miR-7-5p/SQSTM1 signaling pathway was the mechanism underlying low SS inhibiting endothelial mitophagy that involves mitochondrial homeostasis impairment and endothelial dysfunction.
Collapse
Affiliation(s)
- Weike Liu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Huajing Song
- School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Jing Xu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuqi Guo
- School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Chunju Zhang
- School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yanli Yao
- School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Hua Zhang
- School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhendong Liu
- School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Yue-Chun Li
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
11
|
Zhang W, Wang H, Yuan Z, Chu G, Sun H, Yu Z, Liang H, Liu T, Zhou F, Li B. Moderate mechanical stimulation rescues degenerative annulus fibrosus by suppressing caveolin-1 mediated pro-inflammatory signaling pathway. Int J Biol Sci 2021; 17:1395-1412. [PMID: 33867854 PMCID: PMC8040478 DOI: 10.7150/ijbs.57774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Mechanical loading can induce or antagonize the extracellular matrix (ECM) synthesis, proliferation, migration, and inflammatory responses of annulus fibrosus cells (AFCs), depending on the loading mode and level. Caveolin-1 (Cav1), the core protein of caveolae, plays an important role in cellular mechanotransduction and inflammatory responses. In the present study, we presented that AFCs demonstrated different behaviors when subjected to cyclic tensile strain (CTS) for 24 h at a magnitude of 0%, 2%, 5% and 12%, respectively. It was found that 5% CTS had positive effects on cell proliferation, migration and anabolism, while 12% CTS had the opposite effects. Besides, cells exposed to interleukin-1β stimulus exhibited an increase expression in inflammatory genes, and the expression of these genes decreased after exposure to moderate mechanical loading with 5% CTS. In addition, 5% CTS decreased the level of Cav1 and integrin β1 and exhibited anti-inflammatory effects. Moreover, the expression of integrin β1 and p-p65 increased in AFCs transfected with Cav1 plasmids. In vivo results revealed that moderate mechanical stimulation could recover the water content and morphology of the discs. In conclusion, moderate mechanical stimulation restrained Cav1-mediated signaling pathway and exhibited anti-inflammatory effects on AFCs. Together with in vivo results, this study expounds the underlying molecular mechanisms on the effect of moderate mechanical stimulation on intervertebral discs (IVDs) and may provide a new therapeutic strategy for the treatment of IVD degeneration.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Zhangqin Yuan
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Genglei Chu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Heng Sun
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Zilin Yu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Huan Liang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Tao Liu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Feng Zhou
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China.,China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Tanaka K, Joshi D, Timalsina S, Schwartz MA. Early events in endothelial flow sensing. Cytoskeleton (Hoboken) 2021; 78:217-231. [PMID: 33543538 DOI: 10.1002/cm.21652] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Responses of vascular and lymphatic endothelial cells (ECs) to fluid shear stress (FSS) from blood or lymphatic fluid flow govern the development, physiology, and diseases of these structures. Extensive research has characterized the signaling, gene expression and cytoskeletal pathways that mediate effects on EC phenotype and vascular morphogenesis. But the primary mechanisms by which ECs transduce the weak forces from flow into biochemical signals are less well understood. This review covers recent advances in our understanding of the immediate mechanisms of FSS mechanotransduction, integrating results from different disciplines, addressing their roles in development, physiology and disease, and suggesting important questions for future work.
Collapse
Affiliation(s)
- Keiichiro Tanaka
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Divyesh Joshi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Sushma Timalsina
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA.,Department of Cell Biology, Yale University, New Haven, Connecticut, USA.,Department of Biomedical engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Parton RG, Kozlov MM, Ariotti N. Caveolae and lipid sorting: Shaping the cellular response to stress. J Cell Biol 2020; 219:133844. [PMID: 32328645 PMCID: PMC7147102 DOI: 10.1083/jcb.201905071] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Caveolae are an abundant and characteristic surface feature of many vertebrate cells. The uniform shape of caveolae is characterized by a bulb with consistent curvature connected to the plasma membrane (PM) by a neck region with opposing curvature. Caveolae act in mechanoprotection by flattening in response to increased membrane tension, and their disassembly influences the lipid organization of the PM. Here, we review evidence for caveolae as a specialized lipid domain and speculate on mechanisms that link changes in caveolar shape and/or protein composition to alterations in specific lipid species. We propose that high membrane curvature in specific regions of caveolae can enrich specific lipid species, with consequent changes in their localization upon caveolar flattening. In addition, we suggest how changes in the association of lipid-binding caveolar proteins upon flattening of caveolae could allow release of specific lipids into the bulk PM. We speculate that the caveolae-lipid system has evolved to function as a general stress-sensing and stress-protective membrane domain.
Collapse
Affiliation(s)
- Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Australia
| | - Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nicholas Ariotti
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Kensington, Australia.,Department of Pathology, School of Medical Sciences, The University of New South Wales, Kensington, Australia
| |
Collapse
|
14
|
Jalel A, Midani F, Fredj SH, Messaoud T, Hentati F, Soualmia H. Association of BglII Polymorphism in ITGA2 and (894G/T and -786T/C) Polymorphisms in eNOS Gene With Stroke Susceptibility in Tunisian Patients α2 Gene Polymorphism in α2β1 Integrin and eNOS Gene Variants and Stroke. Biol Res Nurs 2020; 23:408-417. [PMID: 33297767 DOI: 10.1177/1099800420977685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND This study investigated the association of BglII polymorphism in α2β1 integrin gene (ITGA2) and eNOS (894G/T and -786T/C) polymorphisms with ischemic stroke (IS) in Tunisian patients. METHODS The study comprised 210 patients with IS and 208 controls. The genotypes of the BglII polymorphism in ITGA2 and eNOS (894G/T and -786T/C) polymorphisms were determined using the PCR-RFLP. The χ2 test was used and the genotype data comparison included heterozygous groups. Haplotype estimation and multiple logistic regression analysis were performed to analyze the significance of polymorphisms. RESULTS The genotype distribution of the BglII polymorphism was significantly different between cases and controls (p < 0.004). This polymorphism was associated with the risk of IS (OR = 3.38, p < 0.001) for the BglII(+/+) genotype. Likewise, the genotype distributions of eNOS (894G/T and -786T/C) polymorphisms were significantly different between the two groups (p < 0.005 and p < 0.01, respectively). The 894G/T polymorphism increased the risk of IS for the TT genotype (OR = 2.23, p < 0.008) and the GT genotype (OR = 1.74, p < 0.009). In addition, the -786T/C variant in the eNOS gene was a risk factor for IS for CC homozygous (OR = 2.52, p < 0.005). T-C Haplotype (OR = 3.06) from combination of the eNOS (894G/T and -786T/C) and T-C-BglII(+) haplotype (OR = 2.76) from combination of eNOS and ITGA2 polymorphisms represented high risks for IS. CONCLUSIONS This study suggests that the BglII variant in ITGA2 is associated with IS susceptibility. Furthermore, the 894G/T and -786T/C polymorphisms in the eNOS gene may be considered as genetic risk factors for IS in the Tunisian population.
Collapse
Affiliation(s)
- Akrem Jalel
- University of Tunis El Manar, High Institute of Medical Technologies of Tunis, Tunisia
| | - Fatma Midani
- University of Tunis El Manar, High Institute of Medical Technologies of Tunis, Tunisia.,University of Carthage, Faculty of Sciences of Bizerte, Tunis, Tunisia
| | - Sondess Hadj Fredj
- University of Tunis El Manar, Research Laboratory "LR99ES11," Biochemistry Laboratory, Children' Hospital, Tunis, Tunisia
| | - Taieb Messaoud
- University of Tunis El Manar, Research Laboratory "LR99ES11," Biochemistry Laboratory, Children' Hospital, Tunis, Tunisia
| | - Fayçal Hentati
- University of Tunis El Manar, Faculty of Medicine, Neuroscience Department, Tunis, Tunisia.,37964University of Tunis El Manar, Neurology Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Hayet Soualmia
- University of Tunis El Manar, High Institute of Medical Technologies of Tunis, Tunisia
| |
Collapse
|
15
|
Wang S, Zhang H, Liu Y, Li L, Guo Y, Jiao F, Fang X, Jefferson JR, Li M, Gao W, Gonzalez-Fernandez E, Maranon RO, Pabbidi MR, Liu R, Alexander BT, Roman RJ, Fan F. Sex differences in the structure and function of rat middle cerebral arteries. Am J Physiol Heart Circ Physiol 2020; 318:H1219-H1232. [PMID: 32216612 PMCID: PMC7346534 DOI: 10.1152/ajpheart.00722.2019] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epidemiological studies demonstrate that there are sex differences in the incidence, prevalence, and outcomes of cerebrovascular disease (CVD). The present study compared the structure and composition of the middle cerebral artery (MCA), neurovascular coupling, and cerebrovascular function and cognition in young Sprague-Dawley (SD) rats. Wall thickness and the inner diameter of the MCA were smaller in females than males. Female MCA exhibited less vascular smooth muscle cells (VSMCs), diminished contractile capability, and more collagen in the media, and a thicker internal elastic lamina with fewer fenestrae compared with males. Female MCA had elevated myogenic tone, lower distensibility, and higher wall stress. The stress/strain curves shifted to the left in female vessels compared with males. The MCA of females failed to constrict compared with a decrease of 15.5 ± 1.9% in males when perfusion pressure was increased from 40 to 180 mmHg. Cerebral blood flow (CBF) rose by 57.4 ± 4.4 and 30.1 ± 3.1% in females and males, respectively, when perfusion pressure increased from 100 to 180 mmHg. The removal of endothelia did not alter the myogenic response in both sexes. Functional hyperemia responses to whisker-barrel stimulation and cognition examined with an eight-arm water maze were similar in both sexes. These results demonstrate that there are intrinsic structural differences in the MCA between sexes, which are associated with diminished myogenic response and CBF autoregulation in females. The structural differences do not alter neurovascular coupling and cognition at a young age; however, they might play a role in the development of CVD after menopause. NEW & NOTEWORTHY Using perfusion fixation of the middle cerebral artery (MCA) in calcium-free solution at physiological pressure and systematically randomly sampling the sections prepared from the same M2 segments of MCA, we found that there are structural differences that are associated with altered cerebral blood flow (CBF) autoregulation but not neurovascular coupling and cognition in young, healthy Sprague-Dawley (SD) rats. Understanding the intrinsic differences in cerebrovascular structure and function in males and females is essential to develop new pharmaceutical treatments for cerebrovascular disease (CVD).
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Longyang Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ya Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Feng Jiao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Neurosurgery, Peking University People's Hospital, Beijing, China
| | - Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua R Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Man Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ezekiel Gonzalez-Fernandez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rodrigo O Maranon
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ruen Liu
- Department of Neurosurgery, Peking University People's Hospital, Beijing, China
| | - Barbara T Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
16
|
Lu T, Chai Q, Jiao G, Wang XL, Sun X, Furuseth JD, Stulak JM, Daly RC, Greason KL, Cha YM, Lee HC. Downregulation of BK channel function and protein expression in coronary arteriolar smooth muscle cells of type 2 diabetic patients. Cardiovasc Res 2020; 115:145-153. [PMID: 29850792 DOI: 10.1093/cvr/cvy137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 05/22/2018] [Indexed: 12/20/2022] Open
Abstract
Aims Type 2 diabetes (T2D) is strongly associated with cardiovascular morbidity and mortality in patients. Vascular large conductance Ca2+-activated potassium (BK) channels, composed of four pore-forming α subunits (BK-α), and four regulatory β1 subunits (BK-β1), are densely expressed in coronary arterial smooth muscle cells (SMCs) and play an important role in regulating vascular tone and myocardial perfusion. However, the role of BK channels in coronary microvascular dysfunction of human subjects with diabetes is unclear. In this study, we examined BK channel function and protein expression, and BK channel-mediated vasodilation in freshly isolated coronary arterioles from T2D patients. Methods and results Atrial tissues were obtained from 16 patients with T2D and 25 matched non-diabetic subjects during cardiopulmonary bypass procedure. Microvessel videomicroscopy and immunoblot analysis were performed in freshly dissected coronary arterioles and inside-out single BK channel currents was recorded in enzymatically isolated coronary arteriolar SMCs. We found that BK channel sensitivity to physiological Ca2+ concentration and voltage was downregulated in the coronary arteriolar SMCs of diabetic patients, compared with non-diabetic controls. BK channel kinetics analysis revealed that there was significant shortening of the mean open time and prolongation of the mean closed time in diabetic patients, resulting in a remarkable reduction of the channel open probability. Functional studies showed that BK channel activation by dehydrosoyasaponin-1 was diminished and that BK channel-mediated vasodilation in response to shear stress was impaired in diabetic coronary arterioles. Immunoblot experiments confirmed that the protein expressions of BK-α and BK-β1 subunits were significantly downregulated, but the ratio of BK-α/BK-β1 was unchanged in the coronary arterioles of T2D patients. Conclusions Our results demonstrated for the first time that BK channel function and BK channel-mediated vasodilation were abnormal in the coronary microvasculature of diabetic patients, due to decreased protein expression and altered intrinsic properties of BK channels.
Collapse
Affiliation(s)
- Tong Lu
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA
| | - Qiang Chai
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA.,Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 89 Jingshi Road, Jinan, Shandong, PR China
| | - Guoqing Jiao
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA.,Department of Cardiovascular Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Road, Wuxi, Jiangsu, PR China
| | - Xiao-Li Wang
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA
| | - Xiaojing Sun
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA
| | - Jonathan D Furuseth
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA
| | - John M Stulak
- Department of Cardiovascular Surgery, Mayo Clinic, 200 First Street, SW., Rochester, Minnesota, USA
| | - Richard C Daly
- Department of Cardiovascular Surgery, Mayo Clinic, 200 First Street, SW., Rochester, Minnesota, USA
| | - Kevin L Greason
- Department of Cardiovascular Surgery, Mayo Clinic, 200 First Street, SW., Rochester, Minnesota, USA
| | - Yong-Mei Cha
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA
| | - Hon-Chi Lee
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW., Rochester, Minnesota, USA
| |
Collapse
|
17
|
Zemskov EA, Lu Q, Ornatowski W, Klinger CN, Desai AA, Maltepe E, Yuan JXJ, Wang T, Fineman JR, Black SM. Biomechanical Forces and Oxidative Stress: Implications for Pulmonary Vascular Disease. Antioxid Redox Signal 2019; 31:819-842. [PMID: 30623676 PMCID: PMC6751394 DOI: 10.1089/ars.2018.7720] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Oxidative stress in the cell is characterized by excessive generation of reactive oxygen species (ROS). Superoxide (O2-) and hydrogen peroxide (H2O2) are the main ROS involved in the regulation of cellular metabolism. As our fundamental understanding of the underlying causes of lung disease has increased it has become evident that oxidative stress plays a critical role. Recent Advances: A number of cells in the lung both produce, and respond to, ROS. These include vascular endothelial and smooth muscle cells, fibroblasts, and epithelial cells as well as the cells involved in the inflammatory response, including macrophages, neutrophils, eosinophils. The redox system is involved in multiple aspects of cell metabolism and cell homeostasis. Critical Issues: Dysregulation of the cellular redox system has consequential effects on cell signaling pathways that are intimately involved in disease progression. The lung is exposed to biomechanical forces (fluid shear stress, cyclic stretch, and pressure) due to the passage of blood through the pulmonary vessels and the distension of the lungs during the breathing cycle. Cells within the lung respond to these forces by activating signal transduction pathways that alter their redox state with both physiologic and pathologic consequences. Future Directions: Here, we will discuss the intimate relationship between biomechanical forces and redox signaling and its role in the development of pulmonary disease. An understanding of the molecular mechanisms induced by biomechanical forces in the pulmonary vasculature is necessary for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christina N Klinger
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, Arizona
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
18
|
Le Roux AL, Quiroga X, Walani N, Arroyo M, Roca-Cusachs P. The plasma membrane as a mechanochemical transducer. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180221. [PMID: 31431176 PMCID: PMC6627014 DOI: 10.1098/rstb.2018.0221] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
Cells are constantly submitted to external mechanical stresses, which they must withstand and respond to. By forming a physical boundary between cells and their environment that is also a biochemical platform, the plasma membrane (PM) is a key interface mediating both cellular response to mechanical stimuli, and subsequent biochemical responses. Here, we review the role of the PM as a mechanosensing structure. We first analyse how the PM responds to mechanical stresses, and then discuss how this mechanical response triggers downstream biochemical responses. The molecular players involved in PM mechanochemical transduction include sensors of membrane unfolding, membrane tension, membrane curvature or membrane domain rearrangement. These sensors trigger signalling cascades fundamental both in healthy scenarios and in diseases such as cancer, which cells harness to maintain integrity, keep or restore homeostasis and adapt to their external environment. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Anabel-Lise Le Roux
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Xarxa Quiroga
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Nikhil Walani
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, Spain
| | - Marino Arroyo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
- Department of Biomedical Sciences, Universitat de Barcelona, Barcelona 08036, Spain
| |
Collapse
|
19
|
Lee KE, Shin SW, Kim GT, Choi JH, Shim EB. Prediction of Plaque Progression in Coronary Arteries Based on a Novel Hemodynamic Index Calculated From Virtual Stenosis Method. Front Physiol 2019; 10:400. [PMID: 31133862 PMCID: PMC6526757 DOI: 10.3389/fphys.2019.00400] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 03/22/2019] [Indexed: 01/07/2023] Open
Abstract
Rationale Predicting the sites in coronary arteries that are susceptible to plaque deposition is essential for the development of clinical treatment strategies and prevention. However, to date, no physiological biomarkers for this purpose have been developed. We hypothesized that the possibility of plaque deposition at a specific site in the coronary artery is associated with wall shear stress (WSS) and fractional flow reserve (FFR). Background and Objective We proposed a new biomarker called the stenosis susceptibility index (SSI) using the FFR and WSS derived using virtual stenosis method. To validate the clinical efficacy of this index, we applied the method to actual pilot clinical cases. This index non-invasively quantifies the vasodilation effects of vascular endothelial cells relative to FFR variation at a specific coronary artery site. Methods and Results Using virtual stenosis method, we computed maximum WSS and FFR according to the variation in stenotic severity at each potential stenotic site and then plotted the variations of maximum WSS (y-axis) and FFR (x-axis). The slope of the graph indicated a site-specific SSI value. Then we determined the most susceptible sites for plaque deposition by comparing SSI values between the potential sites. Applying this method to seven patients revealed 71.4% in per-patient basis analysis 77.8% accuracy in per-vessel basis analysis in percutaneous coronary intervention (PCI) site prediction. Conclusion The SSI index can be used as a predictive biomarker to identify plaque deposition sites. Patients with relatively smaller SSI values also had a higher tendency for myocardial infarction. In conclusion, sites susceptible to plaque deposition can be identified using the SSI index.
Collapse
Affiliation(s)
- Kyung Eun Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, South Korea.,Bio-Convergence Technology Group, Korea Institute of Industrial Technology, Jeju, South Korea
| | - Sung Woong Shin
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, South Korea
| | | | - Jin Ho Choi
- Department of Cardiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Eun Bo Shim
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
20
|
Chen Y, Harty GJ, Zheng Y, Iyer SR, Sugihara S, Sangaralingham SJ, Ichiki T, Grande JP, Lee HC, Wang XL, Burnett JC. CRRL269. Circ Res 2019; 124:1462-1472. [PMID: 30929579 PMCID: PMC6512967 DOI: 10.1161/circresaha.118.314164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RATIONALE Acute kidney injury (AKI) has a high prevalence and mortality in critically ill patients. It is also a powerful risk factor for heart failure incidence driven by hemodynamic changes and neurohormonal activation. However, no drugs have been approved by the Food and Drug Administration. Endogenous pGC-A (particulate guanylyl cyclase A receptor) activators were reported to preserve renal function and improve mortality in AKI patients, although hypotension accompanied by pGC-A activators have limited their therapeutic potential. OBJECTIVE We investigated the therapeutic potential of a nonhypotensive pGC-A activator/designer natriuretic peptide, CRRL269, in a short-term, large animal model of ischemia-induced AKI and also investigated the potential of uCNP (urinary C-type natriuretic peptide) as a biomarker for AKI. METHODS AND RESULTS We first showed that CRRL269 stimulated cGMP generation, suppressed plasma angiotensin II, and reduced cardiac filling pressures without lowering blood pressure in the AKI canine model. We also demonstrated that CRRL269 preserved glomerular filtration rate, increased renal blood flow, and promoted diuresis and natriuresis. Further, CRRL269 reduced kidney injury and apoptosis as evidenced by ex vivo histology and tissue apoptosis analysis. We also showed, compared with native pGC-A activators, that CRRL269 is a more potent inhibitor of apoptosis in renal cells and induced less decreases in intracellular Ca2+ concentration in vascular smooth muscle cells. The renal antiapoptotic effects were at least mediated by cGMP/PKG pathway. Further, CRRL269 inhibited proapoptotic genes expression using a polymerase chain reaction gene array. Additionally, we demonstrated that AKI increased uCNP levels. CONCLUSIONS Our study supports developing CRRL269 as a novel renocardiac protective agent for AKI treatment.
Collapse
Affiliation(s)
- Yang Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
| | - Gail J. Harty
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Ye Zheng
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Seethalakshmi R. Iyer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Shinobu Sugihara
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - S. Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Joseph P. Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Hon-Chi Lee
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Xiao Li Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - John C. Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| |
Collapse
|
21
|
Liu X, Du H, Chai Q, Jia Q, Liu L, Zhao M, Li J, Tang H, Chen W, Zhao L, Fang L, Gao L, Zhao J. Blocking mitochondrial cyclophilin D ameliorates TSH-impaired defensive barrier of artery. Redox Biol 2018; 15:418-434. [PMID: 29353219 PMCID: PMC5975066 DOI: 10.1016/j.redox.2018.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS Endothelial cells (ECs) constitute the defensive barrier of vasculature, which maintains the vascular homeostasis. Mitochondrial oxidative stress (mitoOS) in ECs significantly affects the initiation and progression of vascular diseases. The higher serum thyroid stimulating hormone (TSH) level is being recognized as a nonconventional risk factor responsible for the increased risk of cardiovascular diseases in subclinical hypothyroidism (SCH). However, effects and underlying mechanisms of elevated TSH on ECs are still ambiguous. We sought to investigate whether cyclophilin D (CypD), emerging as a crucial mediator in mitoOS, regulates effects of TSH on ECs. METHODS AND RESULTS SCH patients with TSH > = 10mIU/L showed a positive correlation between serum TSH and endothelin-1 levels. When TSH levels declined to normal in these subjects after levothyroxine therapy, serum endothelin-1 levels were significantly reduced. Supplemented with exogenous thyroxine to keep normal thyroid hormones, thyroid-specific TSH receptor (TSHR)-knockout mice with injection of exogenous TSH exhibited elevated serum TSH levels, significant endothelial oxidative injuries and disturbed endothelium-dependent vasodilation. However, Tshr-/- mice resisted to TSH-impaired vasotonia. We further confirmed that elevated TSH triggered excessive mitochondrial permeability transition pore (mPTP) opening and mitochondrial oxidative damages in mouse aorta, as well as in cultured ECs. Genetic or pharmacological inhibition of CypD (the key regulator for mPTP opening) attenuated TSH-induced mitochondrial oxidative damages and further rescued endothelial functions. Finally, we confirmed that elevated TSH could activate CypD by enhancing CypD acetylation via inhibiting adenosine monophosphate-activated protein kinase/sirtuin-3 signaling pathway in ECs. CONCLUSIONS These findings reveal that elevated TSH triggers mitochondrial perturbations in ECs and provide insights that blocking mitochondrial CypD enhances the defensive ability of ECs under TSH exposure.
Collapse
Affiliation(s)
- Xiaojing Liu
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, United States
| | - Qiang Chai
- Department of Cardiovascular Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250001, China
| | - Qing Jia
- Department of Cardiovascular Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250001, China
| | - Lu Liu
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Meng Zhao
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Jun Li
- Department of Pharmacy, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Hui Tang
- Department of Pharmacy, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Wenbin Chen
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Lifang Zhao
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Li Fang
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Ling Gao
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China; Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China.
| | - Jiajun Zhao
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China.
| |
Collapse
|
22
|
Lu T, Wang XL, Chai Q, Sun X, Sieck GC, Katusic ZS, Lee HC. Role of the endothelial caveolae microdomain in shear stress-mediated coronary vasorelaxation. J Biol Chem 2017; 292:19013-19023. [PMID: 28924052 DOI: 10.1074/jbc.m117.786152] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
In this study, we determined the role of caveolae and the ionic mechanisms that mediate shear stress-mediated vasodilation (SSD). We found that both TRPV4 and SK channels are targeted to caveolae in freshly isolated bovine coronary endothelial cells (BCECs) and that TRPV4 and KCa2.3 (SK3) channels are co-immunoprecipitated by anti-caveolin-1 antibodies. Acute exposure of BCECs seeded in a capillary tube to 10 dynes/cm2 of shear stress (SS) resulted in activation of TRPV4 and SK currents. However, after incubation with HC067047 (TRPV4 inhibitor), SK currents could no longer be activated by SS, suggesting SK channel activation by SS was mediated through TRPV4. SK currents in BCECs were also activated by isoproterenol or by GSK1016790A (TRPV4 activator). In addition, preincubation of isolated coronary arterioles with apamin (SK inhibitor) resulted in a significant diminution of SSD whereas preincubation with HC067047 produced vasoconstriction by SS. Exposure of BCECs to SS (15 dynes/cm2 16 h) enhanced the production of nitric oxide and prostacyclin (PGI2) and facilitated the translocation of TRPV4 to the caveolae. Inhibition of TRPV4 abolished the SS-mediated intracellular Ca2+ ([Ca2+] i ) increase in BCECs. These results indicate a dynamic interaction in the vascular endothelium among caveolae TRPV4 and SK3 channels. This caveolae-TRPV4-SK3 channel complex underlies the molecular and ionic mechanisms that modulate SSD in the coronary circulation.
Collapse
Affiliation(s)
- Tong Lu
- From the Department of Cardiovascular Medicine
| | | | - Qiang Chai
- From the Department of Cardiovascular Medicine.,the Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Science, Jinan 250062, China
| | | | - Garry C Sieck
- Department of Physiology and Biomedical Engineering, and
| | - Zvonimir S Katusic
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota 55905 and
| | - Hon-Chi Lee
- From the Department of Cardiovascular Medicine,
| |
Collapse
|
23
|
Desai AJ, Miller LJ. Changes in the plasma membrane in metabolic disease: impact of the membrane environment on G protein-coupled receptor structure and function. Br J Pharmacol 2017; 175:4009-4025. [PMID: 28691227 DOI: 10.1111/bph.13943] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
Drug development targeting GPCRs often utilizes model heterologous cell expression systems, reflecting an implicit assumption that the membrane environment has little functional impact on these receptors or on their responsiveness to drugs. However, much recent data have illustrated that membrane components can have an important functional impact on intrinsic membrane proteins. This review is directed toward gaining a better understanding of the structure of the plasma membrane in health and disease, and how this organelle can influence GPCR structure, function and regulation. It is important to recognize that the membrane provides a potential mode of lateral allosteric regulation of GPCRs and can affect the effectiveness of drugs and their biological responses in various disease states, which can even vary among individuals across the population. The type 1 cholecystokinin receptor is reviewed as an exemplar of a class A GPCR that is affected in this way by changes in the plasma membrane. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
24
|
Li Y, Wang XL, Sun X, Chai Q, Li J, Thompson B, Shen WK, Lu T, Lee HC. Regulation of vascular large-conductance calcium-activated potassium channels by Nrf2 signalling. Diab Vasc Dis Res 2017; 14:353-362. [PMID: 28429615 DOI: 10.1177/1479164117703903] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BK channels are major ionic determinants of vasodilation. BK channel function is impaired in diabetic vessels due to accelerated proteolysis of its beta-1 (BK-β1) subunits in response to increased oxidative stress. The nuclear factor E2-related factor-2 (Nrf2) signalling pathway has emerged as a master regulator of cellular redox status, and we hypothesized that it plays a central role in regulating BK channel function in diabetic vessels. We found that Nrf2 expression was markedly reduced in db/db diabetic mouse aortas, and this was associated with significant downregulation of BK-β1. In addition, the muscle ring finger protein 1 (MuRF1), a known E-3 ligase targeting BK-β1 ubiquitination and proteasomal degradation, was significantly augmented. These findings were reproduced by knockdown of Nrf2 by siRNA in cultured human coronary artery smooth muscle cells. In contrast, adenoviral transfer of Nrf2 gene in these cells downregulated MuRF1 and upregulated BK-β1 expression. Activation of Nrf2 by dimethyl fumarate preserved BK-β1 expression and protected BK channel and vascular function in db/db coronary arteries. These results indicate that expression of BK-β1 is closely regulated by Nrf2 and vascular BK channel function can be restored by Nrf2 activation. Nrf2 should be considered a novel therapeutic target in the treatment of diabetic vasculopathy.
Collapse
Affiliation(s)
- Yong Li
- 1 Department of Cardiology, Affiliated Wujin Hospital of Jiangsu University, Changzhou, China
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiao-Li Wang
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiaojing Sun
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Qiang Chai
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- 3 Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, P.R. China
| | - Jingchao Li
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- 4 Department of Emergency Medicine, Henan Provincial People's Hospital, Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Benjamin Thompson
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Win-Kuang Shen
- 5 Department of Cardiovascular Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Tong Lu
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hon-Chi Lee
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
25
|
Shyu HY, Chen MH, Hsieh YH, Shieh JC, Yen LR, Wang HW, Cheng CW. Association of eNOS and Cav-1 gene polymorphisms with susceptibility risk of large artery atherosclerotic stroke. PLoS One 2017; 12:e0174110. [PMID: 28346478 PMCID: PMC5367681 DOI: 10.1371/journal.pone.0174110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/03/2017] [Indexed: 01/25/2023] Open
Abstract
Endothelial nitric oxide synthase (eNOS) is localized in caveole and has important effects on caveolar coordination through its interaction with caveolin-1 (Cav-1), which supports normal functioning of vascular endothelial cells. However, the relationship between genotypic polymorphisms of e-NOS and Cav-1 genes and ischemic stroke (IS) remains lesser reported. This hospital-based case-control study aimed to determine the genetic polymorphisms of the eNOS (Glu298Asp) and Cav-1 (G14713A and T29107A) genes in association with susceptibility risk in patients who had suffered from a large artery atherosclerotic (LAA) stroke. Genotyping determination for these variant alleles was performed using the TaqMan assay. The distributions of observed allelic and genotypic frequencies for the polymorphisms were in Hardy-Weinberg equilibrium in healthy controls. The risk for an LAA stroke in the Asp298 variant was 1.72 (95% CI = 1.09–2.75) versus Glu298 of the eNOS. In the GA/AA (rs3807987) variant, it was 1.79 (95% CI = 1.16–2.74) versus GG and in TA/AA (rs7804372) was 1.61 (95% CI = 1.06–2.43) versus TT of the Cav-1, respectively. A tendency toward an increased LAA stroke risk was significant in carriers with the eNOS Glu298Asp variant in conjunction with the G14713 A and T29107A polymorphisms of the Cav-1 (aOR = 2.03, P-trend = 0.002). A synergistic effect between eNOS and Cav-1 polymorphisms on IS risk elevation was significantly influenced by alcohol drinking, heavy cigarette smoking (P-trend<0.01), and hypercholesterolemia (P-trend < 0.001). In conclusion, genotypic polymorphisms of the eNOS Glu298Asp and Cav-1 14713A/29107A polymorphisms are associated with the elevated risk of LAA stroke among Han Chinese in Taiwan.
Collapse
Affiliation(s)
- Hann-Yeh Shyu
- Section of Neurology, Department of Internal Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hua Chen
- Section of Neurology, Department of Internal Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Jia-Ching Shieh
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Ling-Rong Yen
- Section of Neurology, Department of Internal Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
| | - Hsiao-Wei Wang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
26
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 593] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
27
|
Zakeri R, Moulay G, Chai Q, Ogut O, Hussain S, Takahama H, Lu T, Wang XL, Linke WA, Lee HC, Redfield MM. Left Atrial Remodeling and Atrioventricular Coupling in a Canine Model of Early Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2016; 9:CIRCHEARTFAILURE.115.003238. [PMID: 27758811 DOI: 10.1161/circheartfailure.115.003238] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/16/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Left atrial (LA) compliance and contractility influence left ventricular stroke volume. We hypothesized that diminished LA compliance and contractile function occur early during the development of heart failure with preserved ejection fraction (HFpEF) and impair overall cardiac performance. METHODS AND RESULTS Cardiac magnetic resonance imaging, echocardiography, left ventricular and LA pressure-volume studies, and tissue analyses were performed in a model of early HFpEF (elderly dogs, renal wrap-induced hypertension, exogenous aldosterone; n=9) and young control dogs (sham surgery; n=13). Early HFpEF was associated with LA enlargement, cardiomyocyte hypertrophy, and enhanced LA contractile function (median active emptying fraction 16% [95% confidence interval, 13-24]% versus 12 [10-14]%, P=0.008; end-systolic pressure-volume relationship slope 2.4 [1.9-3.2]mm Hg/mL HFpEF versus 1.5 [1.2-2.2]mm Hg/mL controls, P=0.01). However, atrioventricular coupling was impaired and the curvilinear LA end-reservoir pressure-volume relationship was shifted upward/leftward in HFpEF (LA stiffness constant [βLA] 0.16 [0.11-0.18]mm Hg/mL versus 0.06 [0.04-0.10]mm Hg/mL controls; P=0.002), indicating reduced LA compliance. Impaired atrioventricular coupling and lower LA compliance correlated with lower left ventricular stroke volume. Total fibrosis and titin isoform composition were similar between groups; however, titin was hyperphosphorylated in HFpEF and correlated with βLA. LA microvascular reactivity was diminished in HFpEF versus controls. LA microvascular density tended to be lower in HFpEF and inversely correlated with βLA. CONCLUSIONS In early-stage hypertensive HFpEF, LA cardiomyocyte hypertrophy, titin hyperphosphorylation, and microvascular dysfunction occur in association with increased systolic and diastolic LA chamber stiffness, impaired atrioventricular coupling, and decreased left ventricular stroke volume. These data indicate that maladaptive LA remodeling occurs early during HFpEF development, supporting a concept of global myocardial remodeling.
Collapse
Affiliation(s)
- Rosita Zakeri
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L).
| | - Gilles Moulay
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Qiang Chai
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Ozgur Ogut
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Saad Hussain
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Hiroyuki Takahama
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Tong Lu
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Xiao-Li Wang
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Wolfgang A Linke
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Hon-Chi Lee
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Margaret M Redfield
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| |
Collapse
|
28
|
Abstract
Mechanical forces will have been omnipresent since the origin of life, and living organisms have evolved mechanisms to sense, interpret, and respond to mechanical stimuli. The cardiovascular system in general, and the heart in particular, is exposed to constantly changing mechanical signals, including stretch, compression, bending, and shear. The heart adjusts its performance to the mechanical environment, modifying electrical, mechanical, metabolic, and structural properties over a range of time scales. Many of the underlying regulatory processes are encoded intracardially and are, thus, maintained even in heart transplant recipients. Although mechanosensitivity of heart rhythm has been described in the medical literature for over a century, its molecular mechanisms are incompletely understood. Thanks to modern biophysical and molecular technologies, the roles of mechanical forces in cardiac biology are being explored in more detail, and detailed mechanisms of mechanotransduction have started to emerge. Mechano-gated ion channels are cardiac mechanoreceptors. They give rise to mechano-electric feedback, thought to contribute to normal function, disease development, and, potentially, therapeutic interventions. In this review, we focus on acute mechanical effects on cardiac electrophysiology, explore molecular candidates underlying observed responses, and discuss their pharmaceutical regulation. From this, we identify open research questions and highlight emerging technologies that may help in addressing them.
Collapse
Affiliation(s)
- Rémi Peyronnet
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (R.P., P.K.); Departments of Developmental Biology and Internal Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO (J.M.N.); Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Freiburg, Germany (R.P., P.K.)
| | - Jeanne M Nerbonne
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (R.P., P.K.); Departments of Developmental Biology and Internal Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO (J.M.N.); Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Freiburg, Germany (R.P., P.K.)
| | - Peter Kohl
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (R.P., P.K.); Departments of Developmental Biology and Internal Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO (J.M.N.); Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Freiburg, Germany (R.P., P.K.).
| |
Collapse
|
29
|
Hong JH, Kang JW, Kim DK, Baik SH, Kim KH, Shanta SR, Jung JH, Mook-Jung I, Kim KP. Global changes of phospholipids identified by MALDI imaging mass spectrometry in a mouse model of Alzheimer's disease. J Lipid Res 2015; 57:36-45. [PMID: 26538545 DOI: 10.1194/jlr.m057869] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia; however, at the present time there is no disease-modifying drug for AD. There is increasing evidence supporting the role of lipid changes in the process of normal cognitive aging and in the etiology of age-related neurodegenerative diseases. AD is characterized by the presence of intraneuronal protein clusters and extracellular aggregates of β-amyloid (Aβ). Disrupted Aβ kinetics may activate intracellular signaling pathways, including tau hyperphosphorylation and proinflammatory pathways. We analyzed and visualized the lipid profiles of mouse brains using MALDI-TOF MS. Direct tissue analysis by MALDI-TOF imaging MS (IMS) can determine the relative abundance and spatial distribution of specific lipids in different tissues. We used 5XFAD mice that almost exclusively generate and rapidly accumulate massive cerebral levels of Aβ-42 (1). Our data showed changes in lipid distribution in the mouse frontal cortex, hippocampus, and subiculum, where Aβ plaques are first generated in AD. Our results suggest that MALDI-IMS is a powerful tool for analyzing the distribution of various phospholipids and that this application might provide novel insight into the prediction of disease.
Collapse
Affiliation(s)
- Ji Hye Hong
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Jeong Won Kang
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Dong Kyu Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Hoon Baik
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Ho Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Selina Rahman Shanta
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Jae Hun Jung
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Sciences, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
30
|
Powter EE, Coleman PR, Tran MH, Lay AJ, Bertolino P, Parton RG, Vadas MA, Gamble JR. Caveolae control the anti-inflammatory phenotype of senescent endothelial cells. Aging Cell 2015; 14:102-11. [PMID: 25407919 PMCID: PMC4326911 DOI: 10.1111/acel.12270] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2014] [Indexed: 02/03/2023] Open
Abstract
Senescent endothelial cells (EC) have been identified in cardiovascular disease, in angiogenic tumour associated vessels and in aged individuals. We have previously identified a novel anti-inflammatory senescent phenotype of EC. We show here that caveolae are critical in the induction of this anti-inflammatory senescent state. Senescent EC induced by either the overexpression of ARHGAP18/SENEX or by H₂O₂ showed significantly increased numbers of caveolae and associated proteins Caveolin-1, cavin-1 and cavin-2. Depletion of these proteins by RNA interference decreased senescence induced by ARHGAP18 and by H₂O₂. ARHGAP18 overexpression induced a predominantly anti-inflammatory senescent population and depletion of the caveolae-associated proteins resulted in the preferential reduction in this senescent population as measured by neutrophil adhesion and adhesion protein expression after TNFα treatment. In confirmation, EC isolated from the aortas of CAV-1(-/-) mice failed to induce this anti-inflammatory senescent cell population upon expression of ARHGAP18, whereas EC from wild-type mice showed a significant increase. NF-κB is one of the major transcription factors mediating the induction of E-selectin and VCAM-1 expression, adhesion molecules responsible for leucocyte attachment to EC. TNFα-induced activation of NF-κB was suppressed in ARHGAP18-induced senescent EC, and this inhibition was reversed by Caveolin-1 knock-down. Thus, out results demonstrate that an increase in caveolae and its component proteins in senescent ECs is associated with inhibition of the NF-kB signalling pathway and promotion of the anti-inflammatory senescent pathway.
Collapse
Affiliation(s)
- Elizabeth E. Powter
- Centre for the Endothelium Vascular Biology Program Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| | - Paul R. Coleman
- Centre for the Endothelium Vascular Biology Program Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| | - Mai H. Tran
- Centre for the Endothelium Vascular Biology Program Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| | - Angelina J. Lay
- Centre for the Endothelium Vascular Biology Program Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| | - Patrick Bertolino
- Liver Immunology Group Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis The University of Queensland University of St. Lucia Qld 4072Australia
| | - Mathew A. Vadas
- Centre for the Endothelium Vascular Biology Program Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| | - Jennifer R. Gamble
- Centre for the Endothelium Vascular Biology Program Centenary Institute Sydney Australia
- The University of Sydney NSW 2006Australia
| |
Collapse
|
31
|
Wang C, Qu B, Wang Z, Ju J, Wang Y, Wang Z, Cao P, Wang D. Proteomic identification of differentially expressed proteins in vascular wall of patients with ruptured intracranial aneurysms. Atherosclerosis 2015; 238:201-6. [DOI: 10.1016/j.atherosclerosis.2014.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 01/23/2023]
|
32
|
Chai Q, Lu T, Wang XL, Lee HC. Hydrogen sulfide impairs shear stress-induced vasodilation in mouse coronary arteries. Pflugers Arch 2015; 467:329-40. [PMID: 24793048 DOI: 10.1007/s00424-014-1526-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 04/15/2014] [Accepted: 04/16/2014] [Indexed: 01/01/2023]
Abstract
Hydrogen sulfide has emerged as an important endothelium-dependent vasodilator, but its role in shear stress-mediated dilation of coronary arteries is unclear. We examined the role of H2S on shear stress-mediated dilation of isolated mouse coronary arteries. In these vessels, Na2S produced concentration-dependent dilation, which was significantly inhibited by iberiotoxin and by 4-aminopyridine. In addition, BK and Kv currents in mouse coronary smooth muscle cells were directly activated by Na2S, suggesting that H2S produced vasodilation through BK and Kv channel activation. Using a pressure servo controller system, freshly isolated mouse coronary arteries were subjected to physiological levels of shear stress (1 to 25 dynes/cm(2)) and produced graded dilatory responses, but such effects were diminished in the presence of 100 μM Na2S. Pre-incubation with the cystathionine γ-lyase inhibitor, D,L-propargylglycine (PPG), resulted in a paradoxical augmentation of shear stress-mediated vasodilation. However, in the presence of L-NAME or in coronary arteries from eNOS knockout mice, PPG inhibited shear stress-mediated vasodilation, suggesting an interaction between NO and H2S signaling. Na2S inhibited eNOS activity in cultured mouse aortic endothelial cells and reduced the level of phospho-eNOS(serine 1177). These results suggest that both NO and H2S are important shear stress-mediated vasodilators in mouse coronary arteries but there is a complex interaction between these two signaling pathways that results in paradoxical vasoconstrictive effects of H2S through inhibition of NO generation.
Collapse
Affiliation(s)
- Qiang Chai
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA,
| | | | | | | |
Collapse
|
33
|
Zhu J, Ide H, Fu YY, Teichert AM, Kato H, Weisel RD, Maynes JT, Coles JG, Caldarone CA. Losartan ameliorates “upstream” pulmonary vein vasculopathy in a piglet model of pulmonary vein stenosis. J Thorac Cardiovasc Surg 2014; 148:2550-7. [DOI: 10.1016/j.jtcvs.2014.07.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/26/2014] [Accepted: 07/16/2014] [Indexed: 12/29/2022]
|
34
|
ZHOU TIAN, ZHENG YIMING, QIU JUHUI, HU JIANJUN, SUN DAMING, TANG CHAOJUN, WANG GUIXUE. ENDOTHELIAL MECHANOTRANSDUCTION MECHANISMS FOR VASCULAR PHYSIOLOGY AND ATHEROSCLEROSIS. J MECH MED BIOL 2014. [DOI: 10.1142/s0219519414300063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vascular physiology and disease progression, such as atherosclerosis, are mediated by hemodynamic force generated from blood flow. The hemodynamic force exerts on vascular endothelial cells (ECs), which could perceive the mechanical signals and transmit them into cell interior by multiple potential shear sensors, collectively known as mechanotransduction. However, we do not understand completely how these shear-sensitive components orchestrate physiological and atherosclerotic responses to shear stress. In this review, we provide an overview of biomechanical mechanisms underlying vascular physiology and atherosclerotic progression. Additionally, we summarize current evidences to illustrate that atherosclerotic lesions preferentially develop in arterial regions experiencing disturbance in blood flow, during which endothelial dysfunction is the initial event of atherosclerosis, inflammation plays dominant roles in atherosclerotic progression, and angiogenesis emerges as compensatory explanation for atherosclerotic plaque rupture. Especially in the presence of systemic risk factors (e.g., hyperlipidaemia, hypertension and hyperglycemia), the synergy between these systemic risk factors with hemodynamic factors aggravates atherosclerosis by co-stimulating some of these biomechanical events. Given the hemodynamic environment of vasculature, understanding how the rapid shear-mediated signaling, particularly in combination with systemic risk factors, contribute to atherosclerotic progression through endothelial dysfunction, inflammation and angiogenesis helps to elucidate the role for atherogenic shear stress in specifically localizing atherosclerotic lesions in arterial regions with disturbed flow.
Collapse
Affiliation(s)
- TIAN ZHOU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - YIMING ZHENG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - JUHUI QIU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - JIANJUN HU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - DAMING SUN
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - CHAOJUN TANG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - GUIXUE WANG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
35
|
Yi F, Wang H, Chai Q, Wang X, Shen WK, Willis MS, Lee HC, Lu T. Regulation of large conductance Ca2+-activated K+ (BK) channel β1 subunit expression by muscle RING finger protein 1 in diabetic vessels. J Biol Chem 2014; 289:10853-10864. [PMID: 24570002 DOI: 10.1074/jbc.m113.520940] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The large conductance Ca(2+)-activated K(+) (BK) channel, expressed abundantly in vascular smooth muscle cells (SMCs), is a key determinant of vascular tone. BK channel activity is tightly regulated by its accessory β1 subunit (BK-β1). However, BK channel function is impaired in diabetic vessels by increased ubiquitin/proteasome-dependent BK-β1 protein degradation. Muscle RING finger protein 1 (MuRF1), a muscle-specific ubiquitin ligase, is implicated in many cardiac and skeletal muscle diseases. However, the role of MuRF1 in the regulation of vascular BK channel and coronary function has not been examined. In this study, we hypothesized that MuRF1 participated in BK-β1 proteolysis, leading to the down-regulation of BK channel activation and impaired coronary function in diabetes. Combining patch clamp and molecular biological approaches, we found that MuRF1 expression was enhanced, accompanied by reduced BK-β1 expression, in high glucose-cultured human coronary SMCs and in diabetic vessels. Knockdown of MuRF1 by siRNA in cultured human SMCs attenuated BK-β1 ubiquitination and increased BK-β1 expression, whereas adenoviral expression of MuRF1 in mouse coronary arteries reduced BK-β1 expression and diminished BK channel-mediated vasodilation. Physical interaction between the N terminus of BK-β1 and the coiled-coil domain of MuRF1 was demonstrated by pulldown assay. Moreover, MuRF1 expression was regulated by NF-κB. Most importantly, pharmacological inhibition of proteasome and NF-κB activities preserved BK-β1 expression and BK-channel-mediated coronary vasodilation in diabetic mice. Hence, our results provide the first evidence that the up-regulation of NF-κB-dependent MuRF1 expression is a novel mechanism that leads to BK channelopathy and vasculopathy in diabetes.
Collapse
Affiliation(s)
- Fu Yi
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xian 710032, China
| | - Huan Wang
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Qiang Chai
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Xiaoli Wang
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Win-Kuang Shen
- Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona 85259
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Hon-Chi Lee
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Tong Lu
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905.
| |
Collapse
|
36
|
Zhang WZ. An association of metabolic syndrome constellation with cellular membrane caveolae. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2014; 4:23866. [PMID: 24563731 PMCID: PMC3926988 DOI: 10.3402/pba.v4.23866] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 01/19/2023]
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that can predispose an individual to a greater risk of developing type-2 diabetes and cardiovascular diseases. The cluster includes abdominal obesity, dyslipidemia, hypertension, and hyperglycemia - all of which are risk factors to public health. While searching for a link among the aforementioned malaises, clues have been focused on the cell membrane domain caveolae, wherein the MetS-associated active molecules are colocalized and interacted with to carry out designated biological activities. Caveola disarray could induce all of those individual metabolic abnormalities to be present in animal models and humans, providing a new target for therapeutic strategy in the management of MetS.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- CMP Laboratory, Port Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|