1
|
Ferreira G, Cardozo R, Chavarria L, Santander A, Sobrevia L, Chang W, Gundersen G, Nicolson GL. The LINC complex in blood vessels: from physiology to pathological implications in arterioles. J Physiol 2025. [PMID: 39898417 DOI: 10.1113/jp285906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
The LINC (linker of nucleoskeleton and cytoskeleton) complex is a critical component of the cellular architecture that bridges the nucleoskeleton and cytoskeleton and mediates mechanotransduction to and from the nucleus. Though it plays important roles in all blood vessels, it is in arterioles that this complex plays a pivotal role in maintaining endothelial cell integrity, regulating vascular tone, forming new microvessels and modulating responses to mechanical and biochemical stimuli. It is also important in vascular smooth muscle cells and fibroblasts, where it possibly plays a role in the contractile to secretory phenotypic transformation during atherosclerosis and vascular ageing, and in fibroblasts' migration and inflammatory responses in the adventitia. Physiologically, the LINC complex contributes to the stability of arteriolar structure, adaptations to changes in blood flow and injury repair mechanisms. Pathologically, dysregulation or mutations in LINC complex components can lead to compromised endothelial function, vascular remodelling and exacerbation of cardiovascular diseases such as atherosclerosis (arteriolosclerosis). This review summarizes our current understanding of the roles of the LINC complex in cells from arterioles, highlighting its most important physiological functions, exploring its implications for vascular pathology and emphasizing some of its functional characteristics in endothelial cells. By elucidating the LINC complex's role in health and disease, we aim to provide insights that could improve future therapeutic strategies targeting LINC complex-related vascular disorders.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Romina Cardozo
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Luisina Chavarria
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Axel Santander
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Sao Paulo, Brazil
- Faculty of Medicine and Biomedical Sciences, University of Queensland Centre for Clinical Research (UQCCR), University of Queensland, QLD, Herston, Queensland, Australia
- Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Wakam Chang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Gregg Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Garth L Nicolson
- Department of Molecular Pathology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
2
|
Bathrinarayanan PV, Hallam SM, Grover LM, Vigolo D, Simmons MJH. Microfluidics as a Powerful Tool to Investigate Microvascular Dysfunction in Trauma Conditions: A Review of the State-of-the-Art. Adv Biol (Weinh) 2024; 8:e2400037. [PMID: 39031943 DOI: 10.1002/adbi.202400037] [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: 01/21/2024] [Revised: 04/18/2024] [Indexed: 07/22/2024]
Abstract
Skeletal muscle trauma such as fracture or crush injury can result in a life-threatening condition called acute compartment syndrome (ACS), which involves elevated compartmental pressure within a closed osteo-fascial compartment, leading to collapse of the microvasculature and resulting in necrosis of the tissue due to ischemia. Diagnosis of ACS is complex and controversial due to the lack of standardized objective methods, which results in high rates of misdiagnosis/late diagnosis, leading to permanent neuro-muscular damage. ACS pathophysiology is poorly understood at a cellular level due to the lack of physiologically relevant models. In this context, microfluidics organ-on-chip systems (OOCs) provide an exciting opportunity to investigate the cellular mechanisms of microvascular dysfunction that leads to ACS. In this article, the state-of-the-art OOCs designs and strategies used to investigate microvasculature dysfunction mechanisms is reviewed. The differential effects of hemodynamic shear stress on endothelial cell characteristics such as morphology, permeability, and inflammation, all of which are altered during microvascular dysfunction is highlighted. The article then critically reviews the importance of microfluidics to investigate closely related microvascular pathologies that cause ACS. The article concludes by discussing potential biomarkers of ACS with a special emphasis on glycocalyx and providing a future perspective.
Collapse
Affiliation(s)
- P Vasanthi Bathrinarayanan
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - S M Hallam
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
| | - L M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - D Vigolo
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - M J H Simmons
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
| |
Collapse
|
3
|
Espina JA, Cordeiro MH, Milivojevic M, Pajić-Lijaković I, Barriga EH. Response of cells and tissues to shear stress. J Cell Sci 2023; 136:jcs260985. [PMID: 37747423 PMCID: PMC10560560 DOI: 10.1242/jcs.260985] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Shear stress is essential for normal physiology and malignancy. Common physiological processes - such as blood flow, particle flow in the gut, or contact between migratory cell clusters and their substrate - produce shear stress that can have an impact on the behavior of different tissues. In addition, shear stress has roles in processes of biomedical interest, such as wound healing, cancer and fibrosis induced by soft implants. Thus, understanding how cells react and adapt to shear stress is important. In this Review, we discuss in vivo and in vitro data obtained from vascular and epithelial models; highlight the insights these have afforded regarding the general mechanisms through which cells sense, transduce and respond to shear stress at the cellular levels; and outline how the changes cells experience in response to shear stress impact tissue organization. Finally, we discuss the role of shear stress in collective cell migration, which is only starting to be appreciated. We review our current understanding of the effects of shear stress in the context of embryo development, cancer and fibrosis, and invite the scientific community to further investigate the role of shear stress in these scenarios.
Collapse
Affiliation(s)
- Jaime A. Espina
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Marilia H. Cordeiro
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Milan Milivojevic
- Faculty of Technology and Metallurgy, Belgrade University, 11120 Belgrade, Serbia
| | | | - Elias H. Barriga
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| |
Collapse
|
4
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
5
|
Abstract
Despite enormous advances, cardiovascular disorders are still a major threat to global health and are responsible for one-third of deaths worldwide. Research for new therapeutics and the investigation of their effects on vascular parameters is often limited by species-specific pathways and a lack of high-throughput methods. The complex 3-dimensional environment of blood vessels, intricate cellular crosstalks, and organ-specific architectures further complicate the quest for a faithful human in vitro model. The development of novel organoid models of various tissues such as brain, gut, and kidney signified a leap for the field of personalized medicine and disease research. By utilizing either embryonic- or patient-derived stem cells, different developmental and pathological mechanisms can be modeled and investigated in a controlled in vitro environment. We have recently developed self-organizing human capillary blood vessel organoids that recapitulate key processes of vasculogenesis, angiogenesis, and diabetic vasculopathy. Since then, this organoid system has been utilized as a model for other disease processes, refined, and adapted for organ specificity. In this review, we will discuss novel and alternative approaches to blood vessel engineering and explore the cellular identity of engineered blood vessels in comparison to in vivo vasculature. Future perspectives and the therapeutic potential of blood vessel organoids will be discussed.
Collapse
Affiliation(s)
- Kirill Salewskij
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna (K.S., J.M.P.).,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Austria (K.S.)
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna (K.S., J.M.P.).,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada (J.M.P.)
| |
Collapse
|
6
|
Decker KP, Chiu A, Weggen JB, Richardson JW, Hogwood AC, Darling AM, Garten RS. High sodium intake differentially impacts brachial artery dilation when evaluated with reactive versus active hyperemia in salt resistant individuals. J Appl Physiol (1985) 2023; 134:277-287. [PMID: 36548512 DOI: 10.1152/japplphysiol.00461.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This study sought to determine if high sodium (HS) intake in salt resistant (SR) individuals attenuates upper limb arterial dilation in response to reactive (occlusion) and active (exercise) hyperemia, two stimuli with varying vasodilatory mechanisms, and the role of oxidative stress in this response. Ten young, SR participants (9 males, 1 female) consumed a 7-day HS (6,900 mg/day) and a 7-day recommended sodium intake (RI: 2,300 mg/day) diet in a randomized order. On the last day of each diet, brachial artery (BA) function was evaluated via reactive (RH-FMD: 5 min of cuff occlusion) and active [handgrip (HG) exercise] hyperemia after consumption of both placebo (PL) and antioxidants (AO). The HS diet significantly elevated sodium excretion (P < 0.05), but mean arterial blood pressure was unchanged. During the PL condition, the HS diet significantly reduced RH-FMD when compared with RI diet (P = 0.01), but this reduction was significantly restored (P = 0.01) when supplemented with AO (HS + PL: 5.9 ± 3.4; HS + AO: 8.2 ± 2.7; RI + PL: 8.9 ± 4.7; RI + AO: 7.0 ± 2.1%). BA shear-to-dilation slopes, evaluated across all HG exercise workloads, were not significantly different across sodium intervention or AO supplementation. In SR individuals, HS intake impaired BA function when assessed via RH-FMD, but was restored with acute AO consumption suggesting oxidative stress as a contributor to this dysfunction. However, exercise-induced BA dilation was unaltered, potentially implicating an inability of HS intake to influence the mechanisms responsible for effectively maintaining skeletal muscle perfusion during exercise.NEW & NOTEWORTHY This study examined if high sodium (HS) intake in salt resistant (SR) individuals attenuates brachial artery (BA) flow-mediated dilation in response to reactive (occlusion) and active (exercise) hyperemia. In SR individuals, HS intake impaired reactive hyperemia-induced BA dilation, but not exercise-induced BA dilation. This finding suggests that although brachial artery nitric oxide bioavailability may be reduced following HS intake, the redundant mechanisms associated with adequate upper limb blood flow regulation during exercise are maintained.
Collapse
Affiliation(s)
- Kevin P Decker
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Alex Chiu
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Jennifer B Weggen
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Jacob W Richardson
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Austin C Hogwood
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia
| | - Ashley M Darling
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Ryan S Garten
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
7
|
Fazekas-Pongor V, Péterfi A, Major D, Szarvas Z, Fekete M, Tabak AG, Csiszar A, Sonntag WE, Austad SN, Ungvari ZI. Decreased lifespan in female "Munchkin" actors from the cast of the 1939 film version of The Wizard of Oz does not support the hypothesis linking hypopituitary dwarfism to longevity. GeroScience 2022; 44:2527-2539. [PMID: 36334178 PMCID: PMC9768075 DOI: 10.1007/s11357-022-00680-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/21/2022] [Indexed: 11/08/2022] Open
Abstract
In laboratory mice, pituitary dwarfism caused by genetic reduction or elimination of the activity of growth hormone (GH) significantly extends lifespan. The effects of congenital pituitary dwarfism on human longevity are not well documented. To analyse the effects of untreated pituitary dwarfism on human lifespan, the longevity of a diverse group of widely known little people, the 124 adults who played "Munchkins" in the 1939 movie The Wizard of Oz was investigated. Survival of "Munchkin" actors with those of controls defined as cast members of The Wizard of Oz and those of other contemporary Academy Award winning Hollywood movies was compared. According to the Kaplan-Meier survival curves, survival of female and male "Munchkin" actors was shorter than cast controls and Hollywood controls of respective sexes. Cox regression analyses showed that female "Munchkin" actors had significantly higher risk ratios compared to both female cast controls (RR, 1.70; 95% CI, 1.05 to 2.77) and female Hollywood controls (RR, 1.52; 95% CI, 1.03 to 2.24). Similar trends were also discernible for men, albeit point estimates were not significant. The lack of lifespan extension in "Munchkin" actors does not support the hypothesis that hereditary GH deficiency regulates longevity in humans.
Collapse
Affiliation(s)
| | - Anna Péterfi
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Dávid Major
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zsófia Szarvas
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Adam G Tabak
- Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Epidemiology and Public Health, University College London, London, UK
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1313, Oklahoma City, OK, 731042, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Departments of Translational Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - William E Sonntag
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1313, Oklahoma City, OK, 731042, USA
| | - Steven N Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zoltan I Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1313, Oklahoma City, OK, 731042, USA.
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA.
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Departments of Translational Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
8
|
Hua Y, Zhang J, Liu Q, Su J, Zhao Y, Zheng G, Yang Z, Zhuo D, Ma C, Fan G. The Induction of Endothelial Autophagy and Its Role in the Development of Atherosclerosis. Front Cardiovasc Med 2022; 9:831847. [PMID: 35402552 PMCID: PMC8983858 DOI: 10.3389/fcvm.2022.831847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/08/2022] [Indexed: 12/29/2022] Open
Abstract
Increasing attention is now being paid to the important role played by autophagic flux in maintaining normal blood vessel walls. Endothelial cell dysfunction initiates the development of atherosclerosis. In the endothelium, a variety of critical triggers ranging from shear stress to circulating blood lipids promote autophagy. Furthermore, emerging evidence links autophagy to a range of important physiological functions such as redox homeostasis, lipid metabolism, and the secretion of vasomodulatory substances that determine the life and death of endothelial cells. Thus, the promotion of autophagy in endothelial cells may have the potential for treating atherosclerosis. This paper reviews the role of endothelial cells in the pathogenesis of atherosclerosis and explores the molecular mechanisms involved in atherosclerosis development.
Collapse
Affiliation(s)
- Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianqian Liu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Su
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guobin Zheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihui Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Danping Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
9
|
Ishii T, Warabi E, Mann GE. Mechanisms underlying unidirectional laminar shear stress-mediated Nrf2 activation in endothelial cells: Amplification of low shear stress signaling by primary cilia. Redox Biol 2021; 46:102103. [PMID: 34425388 PMCID: PMC8379703 DOI: 10.1016/j.redox.2021.102103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells are sensitive to mechanical stress and respond differently to oscillatory flow versus unidirectional flow. This review highlights the mechanisms by which a wide range of unidirectional laminar shear stress induces activation of the redox sensitive antioxidant transcription factor nuclear factor-E2-related factor 2 (Nrf2) in cultured endothelial cells. We propose that fibroblast growth factor-2 (FGF-2), brain-derived neurotrophic factor (BDNF) and 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) are potential Nrf2 activators induced by laminar shear stress. Shear stress-dependent secretion of FGF-2 and its receptor-mediated signaling is tightly controlled, requiring neutrophil elastase released by shear stress, αvβ3 integrin and the cell surface glycocalyx. We speculate that primary cilia respond to low laminar shear stress (<10 dyn/cm2), resulting in secretion of insulin-like growth factor 1 (IGF-1), which facilitates αvβ3 integrin-dependent FGF-2 secretion. Shear stress induces generation of heparan-binding epidermal growth factor-like growth factor (HB-EGF), which contributes to FGF-2 secretion and gene expression. Furthermore, HB-EGF signaling modulates FGF-2-mediated NADPH oxidase 1 activation that favors casein kinase 2 (CK2)-mediated phosphorylation/activation of Nrf2 associated with caveolin 1 in caveolae. Higher shear stress (>15 dyn/cm2) induces vesicular exocytosis of BDNF from endothelial cells, and we propose that BDNF via the p75NTR receptor could induce CK2-mediated Nrf2 activation. Unidirectional laminar shear stress upregulates gene expression of FGF-2 and BDNF and generation of 15d-PGJ2, which cooperate in sustaining Nrf2 activation to protect endothelial cells against oxidative damage.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
10
|
Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis C, Tousoulis D. Inflammatory Mechanisms Contributing to Endothelial Dysfunction. Biomedicines 2021; 9:781. [PMID: 34356845 PMCID: PMC8301477 DOI: 10.3390/biomedicines9070781] [Citation(s) in RCA: 301] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/30/2021] [Accepted: 07/04/2021] [Indexed: 12/16/2022] Open
Abstract
Maintenance of endothelial cell integrity is an important component of human health and disease since the endothelium can perform various functions including regulation of vascular tone, control of hemostasis and thrombosis, cellular adhesion, smooth muscle cell proliferation, and vascular inflammation. Endothelial dysfunction is encompassed by complex pathophysiology that is based on endothelial nitric oxide synthase uncoupling and endothelial activation following stimulation from various inflammatory mediators (molecular patterns, oxidized lipoproteins, cytokines). The downstream signaling via nuclear factor-κB leads to overexpression of adhesion molecules, selectins, and chemokines that facilitate leukocyte adhesion, rolling, and transmigration to the subendothelial space. Moreover, oscillatory shear stress leads to pro-inflammatory endothelial activation with increased monocyte adhesion and endothelial cell apoptosis, an effect that is dependent on multiple pathways and flow-sensitive microRNA regulation. Moreover, the role of neutrophil extracellular traps and NLRP3 inflammasome as inflammatory mechanisms contributing to endothelial dysfunction has recently been unveiled and is under further investigation. Consequently, and following their activation, injured endothelial cells release inflammatory mediators and enter a pro-thrombotic state through activation of coagulation pathways, downregulation of thrombomodulin, and an increase in platelet adhesion and aggregation owing to the action of von-Willebrand factor, ultimately promoting atherosclerosis progression.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Marios Sagris
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Evangelos Oikonomou
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, University of Athens Medical School, 11527 Athens, Greece
| | - Alexios S. Antonopoulos
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Gerasimos Siasos
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, University of Athens Medical School, 11527 Athens, Greece
| | - Costas Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, University of Athens Medical School, 11527 Athens, Greece; (P.T.); (M.S.); (E.O.); (A.S.A.); (G.S.); (C.T.)
| |
Collapse
|
11
|
Bartosch AMW, Mathews R, Mahmoud MM, Cancel LM, Haq ZS, Tarbell JM. Heparan sulfate proteoglycan glypican-1 and PECAM-1 cooperate in shear-induced endothelial nitric oxide production. Sci Rep 2021; 11:11386. [PMID: 34059731 PMCID: PMC8166914 DOI: 10.1038/s41598-021-90941-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/19/2021] [Indexed: 12/29/2022] Open
Abstract
This study aimed to clarify the role of glypican-1 and PECAM-1 in shear-induced nitric oxide production in endothelial cells. Atomic force microscopy pulling was used to apply force to glypican-1 and PECAM-1 on the surface of human umbilical vein endothelial cells and nitric oxide was measured using a fluorescent reporter dye. Glypican-1 pulling for 30 min stimulated nitric oxide production while PECAM-1 pulling did not. However, PECAM-1 downstream activation was necessary for the glypican-1 force-induced response. Glypican-1 knockout mice exhibited impaired flow-induced phosphorylation of eNOS without changes to PECAM-1 expression. A cooperation mechanism for the mechanotransduction of fluid shear stress to nitric oxide production was elucidated in which glypican-1 senses flow and phosphorylates PECAM-1 leading to endothelial nitric oxide synthase phosphorylation and nitric oxide production.
Collapse
Affiliation(s)
- Anne Marie W Bartosch
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Rick Mathews
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA.,The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Marwa M Mahmoud
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA
| | - Limary M Cancel
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA
| | - Zahin S Haq
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA.
| |
Collapse
|
12
|
Kim B, Arany Z. Could shear stress mimetics delay complications in COVID-19? Trends Cardiovasc Med 2021; 32:71-72. [PMID: 33515686 PMCID: PMC7838584 DOI: 10.1016/j.tcm.2021.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 12/21/2022]
Affiliation(s)
- Boa Kim
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zolt Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
13
|
Hu Y, Chen M, Wang M, Li X. Flow-mediated vasodilation through mechanosensitive G protein-coupled receptors in endothelial cells. Trends Cardiovasc Med 2021; 32:61-70. [PMID: 33406458 DOI: 10.1016/j.tcm.2020.12.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022]
Abstract
Currently, endothelium-dependent vasodilatation involves three main mechanisms: production of nitric oxide (NO) by endothelial nitric oxide synthase (eNOS), synthesis of prostanoids by cyclooxygenase, and/or opening of calcium-sensitive potassium channels. Researchers have proposed multiple mechanosensors that may be involved in flow-mediated vasodilation (FMD), including G protein-coupled receptors (GPCRs), ion channels, and intercellular junction proteins, among others. However, GPCRs are considered the major mechanosensors that play a pivotal role in shear stress signal transduction. Among mechanosensitive GPCRs, G protein-coupled receptor 68, histamine H1 receptors, sphingosine-1-phosphate receptor 1, and bradykinin B2 receptors have been identified as endothelial sensors of flow shear stress regulating flow-mediated vasodilation. Thus, this review aims to expound on the mechanism whereby flow shear stress promotes vasodilation through the proposed mechanosensitive GPCRs in ECs.
Collapse
Affiliation(s)
- Yong Hu
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.247, Beiyuan Street, Jinan, Shandong Province, 250031, China.
| | - Miao Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun, Jilin Province, 130021, China.
| | - Meili Wang
- Department of Obstetrics, Maternal and Child Health Care Hospital of Shandong Province, Shandong University, NO.238, Jingshi East Road, Jinan, Shandong, 250012, China.
| | - Xiucun Li
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.247, Beiyuan Street, Jinan, Shandong Province, 250031, China; Department of Anatomy and Histoembryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, NO.44, Wenhua West Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
14
|
Simonet S, Gosgnach W, Billou L, Lucats L, Royere E, Crespo C, Lapret I, Ragonnet L, Moreau K, Vayssettes-Courchay C, Berson P, Bourguignon MP. GTP-cyclohydrolase deficiency induced peripheral and deep microcirculation dysfunction with age. Microvasc Res 2021; 133:104078. [PMID: 32980388 DOI: 10.1016/j.mvr.2020.104078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023]
Abstract
The present study assessed the impact of impaired tetrahydrobiopterin (BH4) production on vasoreactivity from conduit and small arteries along the vascular tree as seen during aging. For this purpose, the mutant hyperphenylalaninemic mouse (hph-1) was used. This model is reported to be deficient in GTP cyclohydrolase I, a rate limiting enzyme in BH4 biosynthesis. BH4 is a key regulator of vascular homeostasis by regulating the nitric oxide synthase 3 (NOS3) activity. In GTP-CH deficient mice, the aortic BH4 levels were decreased, by -77% in 12 week-middle-aged mice (young) and by -83% in 35-45 week-middle-aged mice (middle-aged). In young hph-1, the mesenteric artery ability to respond to flow was slightly reduced by 9%. Aging induced huge modification in many vascular functions. In middle-aged hph-1, we observed a decrease in aortic cGMP levels, biomarker of NO availability (-46%), in flow-mediated vasodilation of mesenteric artery (-31%), in coronary hyperemia response measured in isolated heart following transient ischemia (-27%) and in cutaneous microcirculation dilation in response to acetylcholine assessed in vivo by laser-doppler technic (-69%). In parallel, the endothelium-dependent relaxation in response to acetylcholine in conduit blood vessel, measured on isolated aorta rings, was unchanged in hph-1 mice whatever the age. Our findings demonstrate that in middle-aged GTP-CH depleted mice, the reduction of BH4 was characterized by an alteration of microcirculation dilatory properties observed in various parts of the vascular tree. Large conduit blood vessels vasoreactivity, ie aorta, was unaltered even in middle-aged mice emphasizing the main BH4-deletion impact on the microcirculation.
Collapse
Affiliation(s)
- Serge Simonet
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Willy Gosgnach
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Lucie Billou
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Laurence Lucats
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Emilie Royere
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Christine Crespo
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Isabelle Lapret
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Lea Ragonnet
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | - Kevin Moreau
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | | | - Pascal Berson
- SERVIER Research Institute, Cardiovascular and Metabolism Discovery Research, Suresnes, France
| | | |
Collapse
|
15
|
Hjelle JT, Miller-Hjelle MA, Dobbie JW. The Biology of the Mesothelium during Peritoneal Dialysis. Perit Dial Int 2020. [DOI: 10.1177/089686089501507s03] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Substantial derangements of mesothelial biology are observed during experimental simulations of dialysis conditions, inferred from the content of human dialysis effluent and visualized by microscopy of human mesothelial biopsies. Canosmotically active solutions be made biocompatible with the osmoregulatory system of the mesothelium? Can the contributions of the mesothelium to host defenses against inflammation and/or infection be supported during CAPD? Do underlying metabolic derangements present in various kidney diseases and end-stage renal disease, regardless of cause, require customized CAPD protocols and solutions? Use of dialysis solutions less directly toxic to the mesothelium is a necessary step toward some day manipulating peritoneal biology by pharmacological and therapeutic modalities.
Collapse
Affiliation(s)
- J. Thomas Hjelle
- Basic Sciences, University of Illinois College of Medicine at Peoria, Peoria, Illinois, U.S.A
| | - Marcia A. Miller-Hjelle
- Basic Sciences, University of Illinois College of Medicine at Peoria, Peoria, Illinois, U.S.A
| | - James W. Dobbie
- The Royal Infirmary, The University of Edinburgh, Edinburgh, Scotland
| |
Collapse
|
16
|
Helix 8 is the essential structural motif of mechanosensitive GPCRs. Nat Commun 2019; 10:5784. [PMID: 31857598 PMCID: PMC6923424 DOI: 10.1038/s41467-019-13722-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
G-protein coupled receptors (GPCRs) are versatile cellular sensors for chemical stimuli, but also serve as mechanosensors involved in various (patho)physiological settings like vascular regulation, cardiac hypertrophy and preeclampsia. However, the molecular mechanisms underlying mechanically induced GPCR activation have remained elusive. Here we show that mechanosensitive histamine H1 receptors (H1Rs) are endothelial sensors of fluid shear stress and contribute to flow-induced vasodilation. At the molecular level, we observe that H1Rs undergo stimulus-specific patterns of conformational changes suggesting that mechanical forces and agonists induce distinct active receptor conformations. GPCRs lacking C-terminal helix 8 (H8) are not mechanosensitive, and transfer of H8 to non-responsive GPCRs confers, while removal of H8 precludes, mechanosensitivity. Moreover, disrupting H8 structural integrity by amino acid exchanges impairs mechanosensitivity. Altogether, H8 is the essential structural motif endowing GPCRs with mechanosensitivity. These findings provide a mechanistic basis for a better understanding of the roles of mechanosensitive GPCRs in (patho)physiology. GPCRs are versatile cellular sensors for chemical stimuli but the molecular mechanisms underlying mechanically induced GPCR activation have remained elusive. Here authors identify the C-terminal helix 8 (H8) as the essential structural motif endowing H1R and other GPCRs with mechanosensitivity.
Collapse
|
17
|
Iring A, Jin YJ, Albarrán-Juárez J, Siragusa M, Wang S, Dancs PT, Nakayama A, Tonack S, Chen M, Künne C, Sokol AM, Günther S, Martínez A, Fleming I, Wettschureck N, Graumann J, Weinstein LS, Offermanns S. Shear stress-induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J Clin Invest 2019; 129:2775-2791. [PMID: 31205027 DOI: 10.1172/jci123825] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 05/01/2019] [Indexed: 12/22/2022] Open
Abstract
Hypertension is a primary risk factor for cardiovascular diseases including myocardial infarction and stroke. Major determinants of blood pressure are vasodilatory factors such as nitric oxide (NO) released from the endothelium under the influence of fluid shear stress exerted by the flowing blood. Several endothelial signaling processes mediating fluid shear stress-induced formation and release of vasodilatory factors have been described. It is, however, still poorly understood how fluid shear stress induces these endothelial responses. Here we show that the endothelial mechanosensitive cation channel PIEZO1 mediated fluid shear stress-induced release of adrenomedullin, which in turn activated its Gs-coupled receptor. The subsequent increase in cAMP levels promoted the phosphorylation of endothelial NO synthase (eNOS) at serine 633 through protein kinase A (PKA), leading to the activation of the enzyme. This Gs/PKA-mediated pathway synergized with the AKT-mediated pathways leading to eNOS phosphorylation at serine 1177. Mice with endothelium-specific deficiency of adrenomedullin, the adrenomedullin receptor, or Gαs showed reduced flow-induced eNOS activation and vasodilation and developed hypertension. Our data identify fluid shear stress-induced PIEZO1 activation as a central regulator of endothelial adrenomedullin release and establish the adrenomedullin receptor and subsequent Gs-mediated formation of cAMP as a critical endothelial mechanosignaling pathway regulating basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Andras Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julián Albarrán-Juárez
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - Péter T Dancs
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Akiko Nakayama
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Anna M Sokol
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| | - Johannes Graumann
- German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
18
|
Kim S, Woo CH. Laminar Flow Inhibits ER Stress-Induced Endothelial Apoptosis through PI3K/Akt-Dependent Signaling Pathway. Mol Cells 2018; 41:964-970. [PMID: 30396238 PMCID: PMC6277562 DOI: 10.14348/molcells.2018.0111] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis preferentially involves in prone area of low and disturbed blood flow while steady and high levels of laminar blood flow are relatively protected from atherosclerosis. Disturbed flow induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). ER stress is caused under stress that disturbs the processing and folding of proteins resulting in the accumulation of misfolded proteins in the ER and activation of the UPR. Prolonged or severe UPR leads to activate apoptotic signaling. Recent studies have indicated that disturbed flow significantly up-regulated p-ATF6α, p-IRE1α, and its target spliced XBP-1. However, the role of laminar flow in ER stress-mediated endothelial apoptosis has not been reported yet. The present study thus investigated the role of laminar flow in ER stress-dependent endothelial cell death. The results demonstrated that laminar flow protects ER stress-induced cleavage forms of PARP-1 and caspase-3. Also, laminar flow inhibits ER stress-induced p-eIF2α, ATF4, CHOP, spliced XBP-1, ATF6 and JNK pathway; these effects are abrogated by pharmacological inhibition of PI3K with wortmannin. Finally, nitric oxide affects thapsigargin-induced cell death in response to laminar flow but not UPR. Taken together, these findings indicate that laminar flow inhibits UPR and ER stress-induced endothelial cell death via PI3K/Akt pathway.
Collapse
Affiliation(s)
- Suji Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415,
Korea
| | - Chang-Hoon Woo
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415,
Korea
| |
Collapse
|
19
|
PAR-1 is a novel mechano-sensor transducing laminar flow-mediated endothelial signaling. Sci Rep 2018; 8:15172. [PMID: 30310081 PMCID: PMC6181929 DOI: 10.1038/s41598-018-33222-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Recent studies have indicated that protease-activated receptor-1 (PAR-1) is involved in cytoprotective and anti-inflammatory responses in endothelial cells (ECs). However, the role of PAR-1 in laminar flow-mediated atheroprotective responses remains unknown. Herein, we investigated whether PAR-1 regulates laminar flow-mediated mechanotransduction in ECs. Confocal analysis showed that PAR-1 was internalized into early endosomes in response to laminar flow. In addition, flow cytometry analysis showed that cell surface expression of PAR-1 was reduced by laminar flow, suggesting that PAR-1 was activated in response to laminar flow. Depletion of PAR-1 using human PAR-1 siRNA inhibited unidirectional laminar flow-mediated actin stress fiber formation and cellular alignment as well as atheroprotective gene expressions in HUVECs. Moreover, PAR-1 knockdown inhibited laminar flow-stimulated eNOS phosphorylation, and inhibited the phosphorylations of Src, AMPK, ERK5 and HDAC5. Furthermore, PAR-1 depletion inhibited laminar flow-mediated anti-inflammatory responses as demonstrated by reduced TNFα-induced VCAM-1 expression and by monocyte adhesion to HUVECs, and prevented laminar flow-mediated anti-apoptotic response. An investigation of the role of PAR-1 in vasomotor modulation using mouse aortic rings revealed that acetylcholine-induced vasorelaxation was diminished in PAR-1 deficient mice compared to littermate controls. Taken together, these findings suggest that PAR-1 be viewed as a novel pharmacologic target for the treatment of vascular diseases, including atherosclerosis.
Collapse
|
20
|
The Role of Endothelial Surface Glycocalyx in Mechanosensing and Transduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1097:1-27. [PMID: 30315537 DOI: 10.1007/978-3-319-96445-4_1] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The endothelial cells (ECs) forming the inner wall of every blood vessel are constantly exposed to the mechanical forces generated by blood flow. The EC responses to these hemodynamic forces play a critical role in the homeostasis of the circulatory system. A variety of mechanosensors and transducers, locating on the EC surface, intra- and trans-EC membrane, and within the EC cytoskeleton, have thus been identified to ensure proper functions of ECs. Among them, the most recent candidate is the endothelial surface glycocalyx (ESG), which is a matrix-like thin layer covering the luminal surface of the EC. It consists of various proteoglycans, glycosaminoglycans, and plasma proteins and is close to other prominent EC mechanosensors and transducers. This chapter summarizes the ESG composition, thickness, and structure observed by different labeling and visualization techniques and in different types of vessels. It also presents the literature in determining the ESG mechanical properties by atomic force microscopy and optical tweezers. The molecular mechanisms by which the ESG plays the role in EC mechanosensing and transduction are described as well as the ESG remodeling by shear stress, the actin cytoskeleton, the membrane rafts, the angiogenic factors, and the sphingosine-1-phosphate.
Collapse
|
21
|
Shear-Induced Nitric Oxide Production by Endothelial Cells. Biophys J 2017; 111:208-21. [PMID: 27410748 DOI: 10.1016/j.bpj.2016.05.034] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/30/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023] Open
Abstract
We present a biochemical model of the wall shear stress-induced activation of endothelial nitric oxide synthase (eNOS) in an endothelial cell. The model includes three key mechanotransducers: mechanosensing ion channels, integrins, and G protein-coupled receptors. The reaction cascade consists of two interconnected parts. The first is rapid activation of calcium, which results in formation of calcium-calmodulin complexes, followed by recruitment of eNOS from caveolae. The second is phosphorylation of eNOS by protein kinases PKC and AKT. The model also includes a negative feedback loop due to inhibition of calcium influx into the cell by cyclic guanosine monophosphate (cGMP). In this feedback, increased nitric oxide (NO) levels cause an increase in cGMP levels, so that cGMP inhibition of calcium influx can limit NO production. The model was used to predict the dynamics of NO production by an endothelial cell subjected to a step increase of wall shear stress from zero to a finite physiologically relevant value. Among several experimentally observed features, the model predicts a highly nonlinear, biphasic transient behavior of eNOS activation and NO production: a rapid initial activation due to the very rapid influx of calcium into the cytosol (occurring within 1-5 min) is followed by a sustained period of activation due to protein kinases.
Collapse
|
22
|
Protein Kinases in Pluripotency—Beyond the Usual Suspects. J Mol Biol 2017; 429:1504-1520. [DOI: 10.1016/j.jmb.2017.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/14/2022]
|
23
|
Suki B, Parameswaran H, Imsirovic J, Bartolák-Suki E. Regulatory Roles of Fluctuation-Driven Mechanotransduction in Cell Function. Physiology (Bethesda) 2017; 31:346-58. [PMID: 27511461 DOI: 10.1152/physiol.00051.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cells in the body are exposed to irregular mechanical stimuli. Here, we review the so-called fluctuation-driven mechanotransduction in which stresses stretching cells vary on a cycle-by-cycle basis. We argue that such mechanotransduction is an emergent network phenomenon and offer several potential mechanisms of how it regulates cell function. Several examples from the vasculature, the lung, and tissue engineering are discussed. We conclude with a list of important open questions.
Collapse
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | | - Jasmin Imsirovic
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | |
Collapse
|
24
|
Laosiripisan J, Parkhurst KL, Tanaka H. Associations of resting heart rate with endothelium-dependent vasodilation and shear rate. Clin Exp Hypertens 2017; 39:150-154. [PMID: 28287891 DOI: 10.1080/10641963.2016.1226890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Heart rate is an independent risk factor for cardiovascular disease and a hemodynamic factor that can modulate blood flow as it affects the frequency of shear stimuli acting on the arterial wall. However, the association between heart rate and endothelium-dependent vasodilation remains highly controversial. We determined the association between heart rate at rest and endothelium-dependent vasodilation in 98 apparently healthy adults (18-63 years). The mild and positive association between heart rate and flow-mediated dilation (FMD) was no longer significant when age and sex or baseline diameter were controlled for. The path analyses revealed that heart rate was not directly related to FMD but the association was indirectly mediated by shear rate, which was confirmed by a bias-corrected bootstrap 95% CIs (0.0157-0.1056). We concluded that even though heart rate and endothelium-dependent vasodilation were associated with shear rate, there was no independent relation between heart rate and FMD.
Collapse
Affiliation(s)
- Jitanan Laosiripisan
- a Cardiovascular Aging Research Laboratory, Department of Kinesiology and Health Education , The University of Texas at Austin , Austin , Texas , USA
| | - Kristin L Parkhurst
- a Cardiovascular Aging Research Laboratory, Department of Kinesiology and Health Education , The University of Texas at Austin , Austin , Texas , USA
| | - Hirofumi Tanaka
- a Cardiovascular Aging Research Laboratory, Department of Kinesiology and Health Education , The University of Texas at Austin , Austin , Texas , USA
| |
Collapse
|
25
|
Wuest SL, Stern P, Casartelli E, Egli M. Fluid Dynamics Appearing during Simulated Microgravity Using Random Positioning Machines. PLoS One 2017; 12:e0170826. [PMID: 28135286 PMCID: PMC5279744 DOI: 10.1371/journal.pone.0170826] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/11/2017] [Indexed: 01/13/2023] Open
Abstract
Random Positioning Machines (RPMs) are widely used as tools to simulate microgravity on ground. They consist of two gimbal mounted frames, which constantly rotate biological samples around two perpendicular axes and thus distribute the Earth's gravity vector in all directions over time. In recent years, the RPM is increasingly becoming appreciated as a laboratory instrument also in non-space-related research. For instance, it can be applied for the formation of scaffold-free spheroid cell clusters. The kinematic rotation of the RPM, however, does not only distribute the gravity vector in such a way that it averages to zero, but it also introduces local forces to the cell culture. These forces can be described by rigid body analysis. Although RPMs are commonly used in laboratories, the fluid motion in the cell culture flasks on the RPM and the possible effects of such on cells have not been examined until today; thus, such aspects have been widely neglected. In this study, we used a numerical approach to describe the fluid dynamic characteristic occurring inside a cell culture flask turning on an operating RPM. The simulations showed that the fluid motion within the cell culture flask never reached a steady state or neared a steady state condition. The fluid velocity depends on the rotational velocity of the RPM and is in the order of a few centimeters per second. The highest shear stresses are found along the flask walls; depending of the rotational velocity, they can reach up to a few 100 mPa. The shear stresses in the "bulk volume," however, are always smaller, and their magnitude is in the order of 10 mPa. In conclusion, RPMs are highly appreciated as reliable tools in microgravity research. They have even started to become useful instruments in new research fields of mechanobiology. Depending on the experiment, the fluid dynamic on the RPM cannot be neglected and needs to be taken into consideration. The results presented in this study elucidate the fluid motion and provide insight into the convection and shear stresses that occur inside a cell culture flask during RPM experiments.
Collapse
Affiliation(s)
- Simon L. Wuest
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Aerospace Biomedical Science and Technology, Space Biology Group, Hergiswil, Switzerland
| | - Philip Stern
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Fluid Mechanics and Hydraulic Machines, Horw, Switzerland
| | - Ernesto Casartelli
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Fluid Mechanics and Hydraulic Machines, Horw, Switzerland
| | - Marcel Egli
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Aerospace Biomedical Science and Technology, Space Biology Group, Hergiswil, Switzerland
- * E-mail:
| |
Collapse
|
26
|
Carneiro AP, Fonseca-Alaniz MH, Dallan LAO, Miyakawa AA, Krieger JE. β-arrestin is critical for early shear stress-induced Akt/eNOS activation in human vascular endothelial cells. Biochem Biophys Res Commun 2017; 483:75-81. [PMID: 28062183 DOI: 10.1016/j.bbrc.2017.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/03/2017] [Indexed: 01/14/2023]
Abstract
Recent evidence suggests that β-arrestins, which are involved in G protein-coupled receptors desensitization, may influence mechanotransduction. Here, we observed that nitric oxide (NO) production was abrogated in human saphenous vein endothelial cells (SVECs) transfected with siRNA against β-arrestin 1 and 2 subjected to shear stress (SS, 15 dynes/cm2, 10 min). The downregulation of β-arrestins 1/2 in SVECs cells also prevented the SS-induced rise in levels of phosphorylation of Akt and endothelial nitric oxide synthase (eNOS, Serine 1177). Interestingly, immunoprecipitation revealed that β-arrestin interacts with Akt, eNOS and caveolin-1 and these interactions are not influenced by SS. Our data indicate that β-arrestins and Akt/eNOS downstream signaling are required for early SS-induced NO production in SVECs, which is consistent with the idea that β-arrestins and caveolin-1 are part of a pre-assembled complex associated with the cellular mechanotransduction machinery.
Collapse
Affiliation(s)
- Ana Paula Carneiro
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | | | - Ayumi Aurea Miyakawa
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Jose Eduardo Krieger
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil.
| |
Collapse
|
27
|
ENDOTHELIUM-DERIVED INHIBITORS EFFICIENTLY ATTENUATE THE AGGREGATION AND ADHESION RESPONSES OF REFRIGERATED PLATELETS. Shock 2016; 45:220-7. [PMID: 26555740 DOI: 10.1097/shk.0000000000000493] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Refrigeration of platelets (4°C) provides the possibility of improving transfusion practice over the current standard-of-care, room temperature (RT) storage. However, the increased level of platelet activation observed at 4°C in vitro is cause for concern of uncontrolled thrombosis in vivo. In this study, we assessed the safety of 4°C-stored platelets by evaluating their response to physiologic inhibitors prostacyclin (PGI2) and nitric oxide (NO). Apheresis platelets were collected from healthy donors (n = 4) and tested on Day 1 (fresh) or Day 5 (RT- and 4°C-stored) after treatment with PGI2 and NO or not for: thrombin generation; factor V (FV) activity; intracellular free calcium, cAMP and cGMP; ATP release; TRAP-induced activation; aggregation to ADP, collagen, and TRAP, and adhesion to collagen under arterial flow. Data were analyzed using two-way ANOVA and post-hoc Tukey test for multiple comparisons, with significance set at P < 0.05. Treatment with inhibitors increased intracellular cAMP and cGMP levels in fresh and stored platelets. Thrombin generation was significantly accelerated in stored platelets consistent with increased factor V levels, PS exposure, CD62P expression, intracellular free calcium, and ATP release. While treatment with inhibitors did not attenuate thrombin generation in stored platelets, activation, aggregation, and adhesion responses were inhibited by both PGI2 and NO in 4°C-stored platelets. In contrast, though RT-stored platelets were activated, they did not adhere or aggregate in response to agonists. Thus, refrigerated platelets maintain their intracellular machinery, are responsive to agonists and platelet function inhibitors, and perform hemostatically better than RT-stored platelets.
Collapse
|
28
|
Dragovich MA, Chester D, Fu BM, Wu C, Xu Y, Goligorsky MS, Zhang XF. Mechanotransduction of the endothelial glycocalyx mediates nitric oxide production through activation of TRP channels. Am J Physiol Cell Physiol 2016; 311:C846-C853. [PMID: 27681180 DOI: 10.1152/ajpcell.00288.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 09/26/2016] [Indexed: 01/28/2023]
Abstract
The endothelial surface glycocalyx (ESG) is a carbohydrate-rich layer found on the vascular endothelium, serving critical functions in the mechanotransduction of blood flow-induced forces. One of the most important protective functions of the ESG is to mediate the production of nitric oxide (NO) in response to blood flow. However, the detailed mechanism underlying ESG's mechanotransduction of the production of NO has not been completely identified. Herein, using the cultured rat brain microvascular endothelial cells (bEnd.3) as a model system, we have implemented a combined atomic force and fluorescence microscopy approach to show that the ESG senses and transduces vertical mechanical stretch to produce NO. This rapid NO production is dependent on the presence of both heparan sulfate (HS) and hyaluronic acid (HA) in ESG, as the removal of HS and/or HA leads to a significant decrease in NO production. Moreover, the production of NO is dependent on the intake of Ca2+ via endothelial transient receptor potential (TRP) channels. Together, our results demonstrate the molecular mechanism of rapid production of NO in response to vertical mechanical stretch.
Collapse
Affiliation(s)
- Matthew A Dragovich
- Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, Pennsylvania
| | - Daniel Chester
- Bioengineering Program, Lehigh University, Bethlehem, Pennsylvania
| | - Bingmei M Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York; and
| | - Chenyu Wu
- Bioengineering Program, Lehigh University, Bethlehem, Pennsylvania
| | - Yan Xu
- Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, Pennsylvania
| | - Michael S Goligorsky
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, New York
| | - X Frank Zhang
- Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, Pennsylvania; .,Bioengineering Program, Lehigh University, Bethlehem, Pennsylvania
| |
Collapse
|
29
|
Andrews AM, Muzorewa TT, Zaccheo KA, Buerk DG, Jaron D, Barbee KA. Cholesterol Enrichment Impairs Capacitative Calcium Entry, eNOS Phosphorylation & Shear Stress-Induced NO Production. Cell Mol Bioeng 2016; 10:30-40. [PMID: 28138348 DOI: 10.1007/s12195-016-0456-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Endothelial dysfunction, characterized by decreased production or availability of nitric oxide (NO), is widely believed to be the hallmark of early-stage atherosclerosis. In addition, hypercholesterolemia is considered a major risk factor for development of atherosclerosis and is associated with impaired flow-induced dilation. However, the mechanism by which elevated cholesterol levels leads to decreased production of NO is unclear. NO is released in response to shear stress and agonist-evoked changes in intracellular calcium. Although calcium signaling is complex, we have previously shown that NO production by endothelial nitric oxide synthase (eNOS) is preferentially activated by calcium influx via store-operated channels. We hypothesized that cholesterol enrichment altered this signaling pathway (known as capacitive calcium entry; CCE) ultimately leading to decreased NO. Our results show that cholesterol enrichment abolished ATP-induced eNOS phosphorylation and attenuated the calcium response by the preferential inhibition of CCE. Furthermore, cholesterol enrichment also inhibited shear stress-induced NO production and eNOS phosporylation, consistent with our previous results showing a significant role for ATP autocrine stimulation and subsequent activation of CCE in the endothelial flow response.
Collapse
Affiliation(s)
- Allison M Andrews
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, 3500N. Broad St., Philadelphia, PA 19140, USA
| | - Tenderano T Muzorewa
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Kelly A Zaccheo
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
30
|
Hsieh ML, Liu PY, Wu JM, Liao JK, Wang JN. Interventional Transcatheter Closure Ameliorates the Leukocyte Rho Kinase Activities among Patients with Patent Ductus Arteriosus. ACTA CARDIOLOGICA SINICA 2016; 31:494-9. [PMID: 27122913 DOI: 10.6515/acs20150424d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Patent ductus arteriosus (PDA) causes increased pulmonary blood flow, which can lead to pulmonary arterial hypertension (PAH). Rho-associated coiled-coil containing protein kinase (ROCK) may play an important pathophysiological role in PAH. We hypothesized that the increased pulmonary artery (PA) flow from PDA could activate ROCK. METHODS Patients who received a PDA transcatheter closure in our hospital were consecutively enrolled in this study. Basic demographics and clinical hemodynamic data of the study participants were recorded. Then, ROCK activity was measured before and after the PDA occlusion procedure. ROCK activity was defined as the phosphorylation ratio of myosin-binding subunit by Western blot measurement. We also sub-divided patients into the coil group and occluder group based on the occlusion device used in each patient's procedure. RESULTS From January 2009 to December 2011, 25 patients with a median age of 2.3 years, ranging from 10 months to 72 years were enrolled. The mean PDA size was 0.31 ± 0.14 cm, the mean Qp/Qs shunt was 1.54 ± 0.41, and the mean systolic pulmonary artery pressure was 26.9 ± 10.3 mmHg. There were 10 patients (one boy and nine girls) in the coil group and 15 patients (four boys and eleven girls) in the occluder group. Following the closure of the PDA, ROCK activity significantly decreased (1.78 ± 2.25 vs. 0.77 ± 0.69, p < 0.01). There was a strong correlation between the leukocyte ROCK activity with the systolic PA pressure (y = 5.4608x + 22.54, R2 = 0.5539, p < 0.05), but not the Qp/Qs value. Both subgroups showed significant changes of ROCK activity after the procedure. Interestingly, when comparing the coil group with the occluder group, the decrease in ROCK activity was more apparent in the occluder group. CONCLUSIONS The findings of this study indicated that ROCK activity is higher in patients with PDA and correlates with PA pressure. The decrease in ROCK activity following the device closure suggests that ROCK may be an important biomarker for PDA patency. KEY WORDS Patent ductus arteriosus (PDA); Pulmonary arterial pressure; Rho kinase; Transcatheter closure.
Collapse
Affiliation(s)
| | - Ping-Yen Liu
- Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University; ; Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - James K Liao
- Division of Cardiology, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
31
|
Al-Khazraji BK, Jackson DN, Goldman D. A Microvascular Wall Shear Rate Function Derived From In Vivo Hemodynamic and Geometric Parameters in Continuously Branching Arterioles. Microcirculation 2016; 23:311-9. [PMID: 27018869 DOI: 10.1111/micc.12279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/26/2016] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Conventional approaches to WSR estimation in the microcirculation involve assumptions that may result in under-/over-estimation of WSR. Therefore, our objectives were: (i) calculate WSR from RBC velocity profiles for a wide range of arteriolar diameters, (ii) provide an experimentally derived and straightforward WSR estimation function, and (iii) compare calculated to conventional WSR estimations. METHODS We characterized RBC velocity profiles in arterioles (n = 39) of branching networks (21-115 μm) in the rat gluteus maximus muscle (n = 6). Measures included mean and maximum velocities, CFL thickness, and RBC column edge velocity, and an experiment-based WSR function was derived. RESULTS CFL thickness (1-4.3 μm) positively correlated with arteriolar diameter (r(2) = 0.64). Results from the WSR equation were similar to values from edge RBC velocities/CFL. Experimental WSRs (1317-4334/sec) were independent of arteriolar diameter, and were greater than pseudoshear rates (for VRatio of 1.6, 2, or diameter-dependent VRatio function) (p < 0.05). CONCLUSION A WSR equation was derived from experimental hemodynamic parameters, and is adaptable to other velocity measurement techniques in order to obtain WSR and stress (when plasma viscosity is known). These findings provide insight on the nature of conventional WSR calculation methods in underestimating microvascular WSR values.
Collapse
Affiliation(s)
- Baraa K Al-Khazraji
- Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada
| | - Dwayne N Jackson
- Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada.,Biomedical Engineering Graduate Program, The University of Western Ontario, London, Ontario, Canada
| | - Daniel Goldman
- Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada.,Biomedical Engineering Graduate Program, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
32
|
The role of endothelial mechanosensitive genes in atherosclerosis and omics approaches. Arch Biochem Biophys 2015; 591:111-31. [PMID: 26686737 DOI: 10.1016/j.abb.2015.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is the leading cause of morbidity and mortality in the U.S., and is a multifactorial disease that preferentially occurs in regions of the arterial tree exposed to disturbed blood flow. The detailed mechanisms by which d-flow induces atherosclerosis involve changes in the expression of genes, epigenetic patterns, and metabolites of multiple vascular cells, especially endothelial cells. This review presents an overview of endothelial mechanobiology and its relation to the pathogenesis of atherosclerosis with special reference to the anatomy of the artery and the underlying fluid mechanics, followed by a discussion of a variety of experimental models to study the role of fluid mechanics and atherosclerosis. Various in vitro and in vivo models to study the role of flow in endothelial biology and pathobiology are discussed in this review. Furthermore, strategies used for the global profiling of the genome, transcriptome, miR-nome, DNA methylome, and metabolome, as they are important to define the biological and pathophysiological mechanisms of atherosclerosis. These "omics" approaches, especially those which derive data based on a single animal model, provide unprecedented opportunities to not only better understand the pathophysiology of atherosclerosis development in a holistic and integrative manner, but also to identify novel molecular and diagnostic targets.
Collapse
|
33
|
Vascular nitric oxide: Beyond eNOS. J Pharmacol Sci 2015; 129:83-94. [PMID: 26499181 DOI: 10.1016/j.jphs.2015.09.002] [Citation(s) in RCA: 534] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/11/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
As the first discovered gaseous signaling molecule, nitric oxide (NO) affects a number of cellular processes, including those involving vascular cells. This brief review summarizes the contribution of NO to the regulation of vascular tone and its sources in the blood vessel wall. NO regulates the degree of contraction of vascular smooth muscle cells mainly by stimulating soluble guanylyl cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP), although cGMP-independent signaling [S-nitrosylation of target proteins, activation of sarco/endoplasmic reticulum calcium ATPase (SERCA) or production of cyclic inosine monophosphate (cIMP)] also can be involved. In the blood vessel wall, NO is produced mainly from l-arginine by the enzyme endothelial nitric oxide synthase (eNOS) but it can also be released non-enzymatically from S-nitrosothiols or from nitrate/nitrite. Dysfunction in the production and/or the bioavailability of NO characterizes endothelial dysfunction, which is associated with cardiovascular diseases such as hypertension and atherosclerosis.
Collapse
|
34
|
Wang S, Iring A, Strilic B, Albarrán Juárez J, Kaur H, Troidl K, Tonack S, Burbiel JC, Müller CE, Fleming I, Lundberg JO, Wettschureck N, Offermanns S. P2Y₂ and Gq/G₁₁ control blood pressure by mediating endothelial mechanotransduction. J Clin Invest 2015; 125:3077-86. [PMID: 26168216 DOI: 10.1172/jci81067] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Elevated blood pressure is a key risk factor for developing cardiovascular diseases. Blood pressure is largely determined by vasodilatory mediators, such as nitric oxide (NO), that are released from the endothelium in response to fluid shear stress exerted by the flowing blood. Previous work has identified several mechanotransduction signaling processes that are involved in fluid shear stress-induced endothelial effects, but how fluid shear stress initiates the response is poorly understood. Here, we evaluated human and bovine endothelial cells and found that the purinergic receptor P2Y2 and the G proteins Gq/G11 mediate fluid shear stress-induced endothelial responses, including [Ca2+]i transients, activation of the endothelial NO synthase (eNOS), phosphorylation of PECAM-1 and VEGFR-2, as well as activation of SRC and AKT. In response to fluid shear stress, endothelial cells released ATP, which activates the purinergic P2Y2 receptor. Mice with induced endothelium-specific P2Y2 or Gq/G11 deficiency lacked flow-induced vasodilation and developed hypertension that was accompanied by reduced eNOS activation. Together, our data identify P2Y2 and Gq/G11 as a critical endothelial mechanosignaling pathway that is upstream of previously described mechanotransduction processes and demonstrate that P2Y2 and Gq/G11 are required for basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
|
35
|
Munn LL. Mechanobiology of lymphatic contractions. Semin Cell Dev Biol 2015; 38:67-74. [PMID: 25636584 DOI: 10.1016/j.semcdb.2015.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 01/30/2023]
Abstract
The lymphatic system is responsible for controlling tissue fluid pressure by facilitating flow of lymph (i.e. the plasma and cells that enter the lymphatic system). Because lymph contains cells of the immune system, its transport is not only important for fluid homeostasis, but also immune function. Lymph drainage can occur via passive flow or active pumping, and much research has identified the key biochemical and mechanical factors that affect output. Although many studies and reviews have addressed how tissue properties and fluid mechanics (i.e. pressure gradients) affect lymph transport [1-3] there is less known about lymphatic mechanobiology. As opposed to passive mechanical properties, mechanobiology describes the active coupling of mechanical signals and biochemical pathways. Lymphatic vasomotion is the result of a fascinating system affected by mechanical forces exerted by the flowing lymph, including pressure-induced vessel stretch and flow-induced shear stresses. These forces can trigger or modulate biochemical pathways important for controlling the lymphatic contractions. Here, I review the current understanding of lymphatic vessel function, focusing on vessel mechanobiology, and summarize the prospects for a comprehensive understanding that integrates the mechanical and biomechanical control mechanisms in the lymphatic system.
Collapse
Affiliation(s)
- Lance L Munn
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, United States.
| |
Collapse
|
36
|
Wu B, Credeur D, Fryer S, Stoner L. The use of shear rate-diameter dose-response curves as an alternative to the flow-mediated dilation test. Med Hypotheses 2015; 84:85-90. [PMID: 25554537 DOI: 10.1016/j.mehy.2014.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 12/09/2014] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
Abstract
The brachial artery flow-mediated dilation test (FMD) is the non-invasive gold-standard used to test endothelial function. Reduced FMD precedes the development of atherosclerosis and provides an early marker for predicting future cardiovascular disease events. Although, this test is of high potential, it is somewhat limited by poor reproducibility. By utilizing hand warming and grip exercise combined with hierarchical linear modeling, shear rate-diameter dose-response curves may provide a novel and more accurate way to assess endothelial function in humans. Shear rate-diameter dose-response curves could potentially improve upon the traditional FMD measurement and serve as a superior clinical and research tool for assessing cardiovascular disease risk in a variety of populations. The current paper presents testable hypotheses and methodology for assessing the validity and reliability of an alternative to the current FMD test.
Collapse
Affiliation(s)
- Brian Wu
- Keck School of Medicine, Los Angeles, CA, United States
| | - Daniel Credeur
- School of Human Performance and Recreation, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Simon Fryer
- School of Sport and Exercise, University of Gloucestershire, United Kingdom
| | - Lee Stoner
- School of Sport and Exercise, Massey University, New Zealand.
| |
Collapse
|
37
|
|
38
|
Zhou J, Li YS, Chien S. Shear stress-initiated signaling and its regulation of endothelial function. Arterioscler Thromb Vasc Biol 2014; 34:2191-8. [PMID: 24876354 DOI: 10.1161/atvbaha.114.303422] [Citation(s) in RCA: 408] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Atherosclerosis develops preferentially at branches and curvatures of the arterial tree, where blood flow pattern is disturbed rather than being laminar, and wall shear stress has an irregular distribution without defined directions. The endothelium in the atherosusceptible regions, in comparison to that in atheroresistant regions, shows activation of proproliferative and proinflammatory gene expressions, reduced production of nitric oxide (NO), increased leukocyte adhesion, and permeability, as well as other atheroprone phenotypes. Differences in gene expressions and cell phenotypes have been detected in endothelia residing in native atherosusceptible and atheroresistant regions of the arteries, or in arteries from animal models with artificial creation of disturbed flow. Similar results have also been shown in in vitro systems that apply controlled shear stresses with or without clear directions to cultured endothelial cells in fluid-dynamically designed flow-loading devices. The available evidence indicates that the coordination of multiple signaling networks, rather than individual separate pathways, links the mechanical signals to specific genetic circuitries in orchestrating the mechanoresponsive networks to evoke comprehensive genetic and functional responses.
Collapse
Affiliation(s)
- Jing Zhou
- From the Department of Physiology and Pathophysiology, Basic Medical College of Peking University, Peking University, Beijing, People's Republic of China (J.Z.); and Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla (J.Z., Y.-S.L., S.C.).
| | - Yi-Shuan Li
- From the Department of Physiology and Pathophysiology, Basic Medical College of Peking University, Peking University, Beijing, People's Republic of China (J.Z.); and Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla (J.Z., Y.-S.L., S.C.)
| | - Shu Chien
- From the Department of Physiology and Pathophysiology, Basic Medical College of Peking University, Peking University, Beijing, People's Republic of China (J.Z.); and Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla (J.Z., Y.-S.L., S.C.).
| |
Collapse
|
39
|
Barvitenko NN, Aslam M, Filosa J, Matteucci E, Nikinmaa M, Pantaleo A, Saldanha C, Baskurt OK. Tissue oxygen demand in regulation of the behavior of the cells in the vasculature. Microcirculation 2014; 20:484-501. [PMID: 23441854 DOI: 10.1111/micc.12052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/19/2013] [Indexed: 12/20/2022]
Abstract
The control of arteriolar diameters in microvasculature has been in the focus of studies on mechanisms matching oxygen demand and supply at the tissue level. Functionally, important vascular elements include EC, VSMC, and RBC. Integration of these different cell types into functional units aimed at matching tissue oxygen supply with tissue oxygen demand is only achieved when all these cells can respond to the signals of tissue oxygen demand. Many vasoactive agents that serve as signals of tissue oxygen demand have their receptors on all these types of cells (VSMC, EC, and RBC) implying that there can be a coordinated regulation of their behavior by the tissue oxygen demand. Such functions of RBC as oxygen carrying by Hb, rheology, and release of vasoactive agents are considered. Several common extra- and intracellular signaling pathways that link tissue oxygen demand with control of VSMC contractility, EC permeability, and RBC functioning are discussed.
Collapse
|
40
|
TAKASE SHINYA, LEROND LAURANCE, BERGAN JOHNJ, SCHMID-SCHÖNBEIN GEERTW. The Inflammatory Reaction During Venous Hypertension in the Rat. Microcirculation 2013. [DOI: 10.1111/j.1549-8719.2000.tb00741.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Adaptation of endothelial cells to physiologically-modeled, variable shear stress. PLoS One 2013; 8:e57004. [PMID: 23457646 PMCID: PMC3573044 DOI: 10.1371/journal.pone.0057004] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 01/18/2013] [Indexed: 12/12/2022] Open
Abstract
Endothelial cell (EC) function is mediated by variable hemodynamic shear stress patterns at the vascular wall, where complex shear stress profiles directly correlate with blood flow conditions that vary temporally based on metabolic demand. The interactions of these more complex and variable shear fields with EC have not been represented in hemodynamic flow models. We hypothesized that EC exposed to pulsatile shear stress that changes in magnitude and duration, modeled directly from real-time physiological variations in heart rate, would elicit phenotypic changes as relevant to their critical roles in thrombosis, hemostasis, and inflammation. Here we designed a physiological flow (PF) model based on short-term temporal changes in blood flow observed in vivo and compared it to static culture and steady flow (SF) at a fixed pulse frequency of 1.3 Hz. Results show significant changes in gene regulation as a function of temporally variable flow, indicating a reduced wound phenotype more representative of quiescence. EC cultured under PF exhibited significantly higher endothelial nitric oxide synthase (eNOS) activity (PF: 176.0±11.9 nmol/105 EC; SF: 115.0±12.5 nmol/105 EC, p = 0.002) and lower TNF-a-induced HL-60 leukocyte adhesion (PF: 37±6 HL-60 cells/mm2; SF: 111±18 HL-60/mm2, p = 0.003) than cells cultured under SF which is consistent with a more quiescent anti-inflammatory and anti-thrombotic phenotype. In vitro models have become increasingly adept at mimicking natural physiology and in doing so have clarified the importance of both chemical and physical cues that drive cell function. These data illustrate that the variability in metabolic demand and subsequent changes in perfusion resulting in constantly variable shear stress plays a key role in EC function that has not previously been described.
Collapse
|
42
|
Gazit AZ, Canter CE. Impact of pulmonary vascular resistances in heart transplantation for congenital heart disease. Curr Cardiol Rev 2013; 7:59-66. [PMID: 22548028 PMCID: PMC3197090 DOI: 10.2174/157340311797484213] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 05/23/2011] [Accepted: 06/27/2011] [Indexed: 02/02/2023] Open
Abstract
Congenital heart disease is one of the major diagnoses in pediatric heart transplantation recipients of all age groups. Assessment of pulmonary vascular resistance in these patients prior to transplantation is crucial to determine their candidacy, however, it is frequently inaccurate because of their abnormal anatomy and physiology. This problem places them at significant risk for pulmonary hypertension and right ventricular failure post transplantation. The pathophysiology of pulmonary vascular disease in children with congenital heart disease depends on their pulmonary blood flow patterns, systemic ventricle function, as well as semilunar valves and atrioventricular valves structure and function. In our review we analyze the pathophysiology of pulmonary vascular disease in children with congenital heart disease and end-stage heart failure, and outline the state of the art pre-transplantation medical and surgical management to achieve reverse remodeling of the pulmonary vasculature by using pulmonary vasodilators and mechanical circulatory support.
Collapse
Affiliation(s)
- Avihu Z Gazit
- Divisions of Critical Care and Cardiology, Department of Pediatrics, Saint Louis Children's Hospital, Washington University in Saint Louis, Missouri, USA.
| | | |
Collapse
|
43
|
Stoner L, McCully KK. Peak and time-integrated shear rates independently predict flow-mediated dilation. JOURNAL OF CLINICAL ULTRASOUND : JCU 2012; 40:341-351. [PMID: 22407951 DOI: 10.1002/jcu.21900] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 01/23/2012] [Indexed: 05/31/2023]
Abstract
BACKGROUND To determine whether peak and time-integrated shear rates independently predict flow-mediated dilation (FMD). METHODS Eleven physically active (25 ± 5 years old) male subjects were tested. FMD was defined as the shear rate-diameter relationship. Hierarchical linear modeling was used to estimate brachial artery diameter change with repeated measures of shear rate nested within each subject. Two models were tested: 1) FMD was induced using ischemia-induced hyperemia (2, 4, 6, and 10 minutes); and 2) FMD was induced following transient (ischemia) and steady-state (forearm heating and handgrip exercise) increases in shear rate. For both models we determined whether peak, in addition to time-integrated shear rates, explained a significant portion of variation for diameter change. RESULTS Model 1: Time integrated shear rates explained most of the variation for diameter change. However, peak shear rate explained an additional significant portion of variation. Model 2: The transient condition resulted in significantly (p = 0.012) smaller diameter change per shear rate change than the steady-state condition. However, when specifying peak shear rate as a covariate, the difference between conditions became nonsignificant (p = 0.138). CONCLUSIONS Peak and time-integrated shear rates independently predict FMD. Future studies using the FMD test should consider both parameters.
Collapse
Affiliation(s)
- Lee Stoner
- Department of Kinesiology, University of Georgia, Ramsey Center, Athens, GA 30602, USA
| | | |
Collapse
|
44
|
Melchior B, Frangos JA. Gαq/11-mediated intracellular calcium responses to retrograde flow in endothelial cells. Am J Physiol Cell Physiol 2012; 303:C467-73. [PMID: 22700794 DOI: 10.1152/ajpcell.00117.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Disturbed flow patterns, including reversal in flow direction, are key factors in the development of dysfunctional endothelial cells (ECs) and atherosclerotic lesions. An almost immediate response of ECs to fluid shear stress is the increase in cytosolic calcium concentration ([Ca(2+)](i)). Whether the source of [Ca(2+)](i) is extracellular, released from Ca(2+) intracellular stores, or both is still undefined, though it is likely dependent on the nature of forces involved. We have previously shown that a change in flow direction (retrograde flow) on a flow-adapted endothelial monolayer induces the remodeling of the cell-cell junction along with a dramatic [Ca(2+)](i) burst compared with cells exposed to unidirectional or orthograde flow. The heterotrimeric G protein-α q and 11 subunit (Gα(q/11)) is a likely candidate in effecting shear-induced increases in [Ca(2+)](i) since its expression is enriched at the junction and has been previously shown to be activated within seconds after onset of flow. In flow-adapted human ECs, we have investigated to what extent the Gα(q/11) pathway mediates calcium dynamics after reversal in flow direction. We observed that the elapsed time to peak [Ca(2+)](i) response to a 10 dyn/cm(2) retrograde shear stress was increased by 11 s in cells silenced with small interfering RNA directed against Gα(q/11). A similar lag in [Ca(2+)](i) transient was observed after cells were treated with the phospholipase C (PLC)-βγ inhibitor, U-73122, or the phosphatidylinositol-specific PLC inhibitor, edelfosine, compared with controls. Lower levels of inositol 1,4,5-trisphosphate accumulation seconds after the onset of flow correlated with the increased lag in [Ca(2+)](i) responses observed with the different treatments. In addition, inhibition of the inositol 1,4,5-trisphosphate receptor entirely abrogated flow-induced [Ca(2+)](i). Taken together, our results identify the Gα(q/11)-PLC pathway as the initial trigger for retrograde flow-induced endoplasmic reticulum calcium store release, thereby offering a novel approach to regulating EC dysfunctions in regions subjected to the reversal of blood flow.
Collapse
Affiliation(s)
- Benoît Melchior
- La Jolla Bioengineering Institute, San Diego, California, USA
| | | |
Collapse
|
45
|
Stoner L, McCully KK. Velocity acceleration as a determinant of flow-mediated dilation. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:580-592. [PMID: 22342687 DOI: 10.1016/j.ultrasmedbio.2011.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 12/09/2011] [Accepted: 12/28/2011] [Indexed: 05/31/2023]
Abstract
Shear stress is the established stimulus for flow-mediated dilation (FMD). In vivo, shear stress is typically estimated using mean blood velocity. However, mean blood velocity may not adequately characterize the shear stimulus. Pulsatile flow results in large shear gradients (velocity acceleration) at the onset of flow. The purpose of this study was to determine the importance of velocity acceleration to FMD. We define FMD as the brachial artery shear rate-diameter slope. Fourteen physically active, young (26 ± 5 years), male subjects were tested. Progressive forearm heating and handgrip exercise elicited steady-state increases in shear rate. FMD was measured prior to and following induced increases in velocity acceleration. Velocity acceleration was increased by inflating a tourniquet around the forearm to 40 mm Hg. Hierarchical linear modeling was used to estimate change in diameter with repeated measures of shear stress nested within each subject. Averaged across conditions, the 40 mm Hg cuff resulted in a 14% increase in velocity acceleration (p = 0.001). FMD was attenuated by 11.0% (p = 0.015) for the acceleration vs. control condition. However, after specifying velocity acceleration as a covariate, FMD was no longer significantly (p = 0.619) different between acceleration and control conditions. This finding suggests that mean blood velocity alone may not adequately characterize the shear stimulus.
Collapse
Affiliation(s)
- Lee Stoner
- School of Sport and Exercise, Massey University, Wellington, New Zealand.
| | | |
Collapse
|
46
|
Nunez F, Trach S, Burnett L, Handa R, Van Dyke M, Callahan M, Smith T. Vasoactive properties of keratin-derived compounds. Microcirculation 2012; 18:663-9. [PMID: 21977948 DOI: 10.1111/j.1549-8719.2011.00135.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Keratin proteins have been utilized as biomaterials for decades, and are currently under investigation for a variety of tissue regeneration and trauma applications. It has been suggested that certain keratins may have the capacity to act as a colloid in fluid resuscitation applications, providing viscosity and oncotic properties that may be beneficial during acute ischemic events. Oxidized keratin derivatives, also known as keratoses, show good blood and cardiovascular compatibility and thus are the subject of this study. METHODS The effects of keratose compounds will be assessed using a topload i.v. infusion model and observation of changes in the microvasculature of the cremaster muscle of rats. RESULTS Keratose resuscitation fluid (KRF) administration resulted in significant vasodilation in the cremaster muscle. This effect was blocked with pretreatment of l-NA to inhibit NO. Another keratin fraction, alpha-keratose, which is the primary viscosic compound, was not found to induce vasodilation. CONCLUSIONS The apparent mechanism of vasodilation was found to be NO-mediated and isolated to a particular purified fraction, the KAP.
Collapse
Affiliation(s)
- Fiesky Nunez
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Medical Center Boulevard, Winston Salem, North Carolina 27157, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Stoner L, Sabatier MJ. Use of ultrasound for non-invasive assessment of flow-mediated dilation. J Atheroscler Thromb 2012; 19:407-21. [PMID: 22659525 DOI: 10.5551/jat.11395] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The pathological complications of atherosclerosis, namely heart attacks and strokes, remain the leading cause of mortality in the Western world. Preceding atherosclerosis is endothelial dysfunction. There is therefore interest in the application of non-invasive clinical tools to assess endothelial function. The flow-mediated dilation (FMD) test is the standard tool used to assess endothelial function. Reduced FMD is an early marker of atherosclerosis and has been noted for its capacity to predict future cardiovascular disease events. This review discusses the measurement of endothelial function using ultrasound, with a focus on the FMD technique.
Collapse
Affiliation(s)
- Lee Stoner
- School of Sport and Exercise, Massey University, Wellington, New Zealand.
| | | |
Collapse
|
48
|
The importance of velocity acceleration to flow-mediated dilation. Int J Vasc Med 2012; 2012:589213. [PMID: 22315688 PMCID: PMC3270398 DOI: 10.1155/2012/589213] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 10/12/2011] [Indexed: 01/22/2023] Open
Abstract
The validity of the flow-mediated dilation test has been questioned due to the lack of normalization to the primary stimulus, shear stress. Shear stress can be calculated using Poiseuille's law. However, little attention has been given to the most appropriate blood velocity parameter(s) for calculating shear stress. The pulsatile nature of blood flow exposes the endothelial cells to two distinct shear stimuli during the cardiac cycle: a large rate of change in shear at the onset of flow (velocity acceleration), followed by a steady component. The parameter typically entered into the Poiseuille's law equation to determine shear stress is time-averaged blood velocity, with no regard for flow pulsatility. This paper will discuss (1) the limitations of using Posieuille's law to estimate shear stress and (2) the importance of the velocity profile-with emphasis on velocity acceleration-to endothelial function and vascular tone.
Collapse
|
49
|
CHIU JENGJIANN, LEE PEILING, USAMI SHUNICHI, CHIEN SHU. DIFFERENTIAL EFFECTS OF SHEAR STRESS ON THE EXPRESSIONS OF ICAM-1 AND VCAM-1 INDUCED BY TNF-α IN ENDOTHELIAL CELLS. J MECH MED BIOL 2011. [DOI: 10.1142/s0219519405001321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vascular endothelial cells (ECs) are subjected to shear stress and cytokine stimulation. We studied the interplay between shear stress and cytokine in modulating the expression of adhesion molecule genes and the adhesive function of ECs. Shear stress (20 dynes/cm2) was applied to ECs prior to or following the addition of tumor necrosis factor (TNF)-α. Shear stress increased the TNF-α-induced expression of intercellular adhesion molecule-1 (ICAM-1) at both mRNA and surface protein levels, but decreased the TNF-α-induced expression of vascular adhesion molecule-1 (VCAM-1). The TNF-α-induced increase in EC adhesiveness for monocytic THP-1 cells was reduced by shear stress. After 24-h pre-shearing followed by 1 h of static incubation, the effect of pre-shearing on TNF-α-induced ICAM-1 mRNA expression vanished. The recovery of the TNF-α-induced VCAM-1 mRNA expression following pre-shearing, however, required a static incubation time of >6 h (completely recovery at 24 h). Pre- and post-shearing caused a reduction in the nuclear factor (NF)-κB-DNA binding activity induced by TNF-α in the EC nucleus. Our findings suggest that shear stress plays differential roles in modulating the TNF-α-induced EC expressions of ICAM-1 and VCAM-1 genes, which serve similar functions in vascular biology.
Collapse
Affiliation(s)
- JENG-JIANN CHIU
- Division of Medical Engineering Research, National Health Research Institutes, Taipei 114, Taiwan, R.O.C
- Institute of Biomedical Engineering, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - PEI-LING LEE
- Division of Medical Engineering Research, National Health Research Institutes, Taipei 114, Taiwan, R.O.C
| | - SHUNICHI USAMI
- Department of Bioengineering and Whitaker Institute of Biomedical Engineering, University of California, San Diego, La Jolla, CA 92093-0427, USA
| | - SHU CHIEN
- Department of Bioengineering and Whitaker Institute of Biomedical Engineering, University of California, San Diego, La Jolla, CA 92093-0427, USA
| |
Collapse
|
50
|
Ahmann KA, Johnson SL, Hebbel RP, Tranquillo RT. Shear stress responses of adult blood outgrowth endothelial cells seeded on bioartificial tissue. Tissue Eng Part A 2011; 17:2511-21. [PMID: 21599543 DOI: 10.1089/ten.tea.2011.0055] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human blood outgrowth endothelial cells (HBOECs) are expanded from circulating endothelial progenitor cells in peripheral blood and thus could provide a source of autologous endothelial cells for tissue-engineered vascular grafts. To examine the suitability of adult HBOECs for use in vascular tissue engineering, the shear stress responsiveness of these cells was examined on bioartificial tissue formed from dermal fibroblasts entrapped in tubular fibrin gels. HBOECs adhered to this surface, deposited collagen IV and laminin, and remained adherent when exposed to 15 dyn/cm(2) shear stress for 24 h. The shear stress responses of HBOECs were compared to human umbilical vein endothelial cells (HUVECs). As with HUVECs, HBOECs upregulated vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 when exposed to tumor necrosis factor (TNF)-α and shear stress decreased the expression of these adhesion molecules on TNF-α-activated monolayers. Nitric oxide production was elevated by shear stress, but did not vary between cell types. Both cell types decreased platelet adhesion to the bioartificial tissue, whereas pre-exposing the cells to flow decreased platelet adhesion further. These results illustrate the potential utility for HBOECs in vascular tissue engineering, as not only do the cells adhere to bioartificial tissue and remain adherent under physiological shear stress, they are also responsive to shear stress signaling.
Collapse
Affiliation(s)
- Katherine A Ahmann
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|