1
|
Zhou Z, Korteland SA, Tardajos-Ayllon B, Wu J, Chambers E, Weninck J, Simons M, Dunning M, Schenkel T, Diagbouga M, Wentzel J, Fragiadaki M, Evans PC. Shear stress is uncoupled from atheroprotective KLK10 in atherosclerotic plaques. Atherosclerosis 2024; 398:118622. [PMID: 39413592 DOI: 10.1016/j.atherosclerosis.2024.118622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND AND AIMS Physiological shear stress promotes vascular homeostasis by inducing protective molecules in endothelial cells (EC). However, physiological shear stress has been linked to atherosclerosis progression in some individuals with heightened cardiovascular risk. To address this apparent paradox, we hypothesized that diseased arteries may exhibit reduced responsiveness to the protective effects of physiological shear stress. Consequently, we compared the transcriptome of EC exposed to physiological shear stress in healthy arteries versus atherosclerotic conditions. METHODS Employing 3D light sheet imaging and computational fluid dynamics, we identified NOS3 as a marker of physiological shear stress in both healthy and atherosclerotic murine arteries. Single-cell RNA sequencing was performed on EC from healthy (C57BL/6) mice, mildly diseased (Apoe-/- normal diet) mice, and highly diseased (Apoe-/- high fat diet) mice. The transcriptomes of Nos3high cells (exposed to physiological shear stress) were compared among the groups. RESULTS Nos3high EC were associated with several markers of physiological shear stress in healthy arteries. Clustering of Nos3high EC revealed 8 different EC subsets that varied in proportion between healthy and diseased arteries. Cluster-specific nested functional enrichment of gene ontology terms revealed that Nos3high EC in diseased arteries were enriched for inflammatory and apoptotic gene expression. These alterations were accompanied by changes in several mechanoreceptors, including the atheroprotective factor KLK10, which was enriched in Nos3high EC in healthy arteries but markedly reduced in severely diseased arteries. CONCLUSIONS Physiological shear stress is uncoupled from atheroprotective KLK10 within atherosclerotic plaques. This sheds light on the complex interplay between shear stress, endothelial function, and the progression of atherosclerosis in individuals at risk of cardiovascular complications.
Collapse
Affiliation(s)
- Ziqi Zhou
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Suze-Anne Korteland
- Department of Cardiology, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Blanca Tardajos-Ayllon
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK; Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Junxi Wu
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Emily Chambers
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Julia Weninck
- Department of Cardiology, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Michael Simons
- Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, CT, United States
| | - Mark Dunning
- Sheffield Bioinformatics Core, The Medical School, University of Sheffield, Sheffield, UK
| | - Torsten Schenkel
- Department of Engineering and Mathematics, Sheffield Hallam University, Sheffield, UK
| | - Mannekomba Diagbouga
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK; Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jolanda Wentzel
- Department of Cardiology, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Maria Fragiadaki
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK; Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
2
|
Vanaei S, Hashemi M, Solouk A, Asghari Ilani M, Amili O, Hefzy MS, Tang Y, Elahinia M. Manufacturing, Processing, and Characterization of Self-Expanding Metallic Stents: A Comprehensive Review. Bioengineering (Basel) 2024; 11:983. [PMID: 39451359 PMCID: PMC11505524 DOI: 10.3390/bioengineering11100983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 10/26/2024] Open
Abstract
This paper aims to review the State of the Art in metal self-expanding stents made from nitinol (NiTi), showing shape memory and superelastic behaviors, to identify the challenges and the opportunities for improving patient outcomes. A significant contribution of this paper is its extensive coverage of multidisciplinary aspects, including design, simulation, materials development, manufacturing, bio/hemocompatibility, biomechanics, biomimicry, patency, and testing methodologies. Additionally, the paper offers in-depth insights into the latest practices and emerging trends, with a special emphasis on the transformative potential of additive manufacturing techniques in the development of metal stents. By consolidating existing knowledge and highlighting areas for future innovation, this review provides a valuable roadmap for advancing nitinol stents.
Collapse
Affiliation(s)
- Saeedeh Vanaei
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| | - Mahdi Hashemi
- Department of Materials and Metallurgical Engineering, Amirkabir University of Technology, Tehran 1591634311, Iran;
| | - Atefeh Solouk
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran;
| | - Mohsen Asghari Ilani
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran 1439957131, Iran;
| | - Omid Amili
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| | - Mohamed Samir Hefzy
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| | - Yuan Tang
- Department of Bioengineering, University of Toledo, Toledo, OH 43606, USA;
| | - Mohammad Elahinia
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| |
Collapse
|
3
|
Panayotov P, Mileva N, Vassilev D. Current Challenges in Coronary Bifurcation Interventions. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1439. [PMID: 39336480 PMCID: PMC11434469 DOI: 10.3390/medicina60091439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024]
Abstract
Coronary bifurcation lesions account for a significant proportion of all percutaneous coronary interventions (PCIs). Interventional treatment of coronary bifurcations is related to significant technical challenges, high complication rates, and worse angiographic and long-term clinical outcomes. This review covers the specific features and structure of coronary bifurcation and explores the main challenges in the interventional treatment of these lesions. This review evaluates various methodologies designed to address these lesions, considering factors such as plaque distribution and bifurcation geometry. It also emphasizes the limitations associated with current techniques. A novel combined optimization approach applied in the interventional treatment of coronary bifurcation may offer superior procedural and long-term outcomes. This combined technique could potentially address the drawbacks of each method, providing a more effective solution for optimizing stent placement in bifurcation lesions. Refining and evaluating these combined techniques is essential for improving clinical outcomes in patients with bifurcation lesions.
Collapse
Affiliation(s)
- Panayot Panayotov
- Department of Cardiology, Pulmonology and Endocrinology, Medical Faculty, Medical University of Pleven, 5800 Pleven, Bulgaria
- Medica Cor Hospital, 7013 Ruse, Bulgaria
| | | | - Dobrin Vassilev
- Medica Cor Hospital, 7013 Ruse, Bulgaria
- Faculty of Public Health and Healthcare, Ruse University "Angel Kanchev", 7017 Ruse, Bulgaria
| |
Collapse
|
4
|
Vuong TNAM, Bartolf‐Kopp M, Andelovic K, Jungst T, Farbehi N, Wise SG, Hayward C, Stevens MC, Rnjak‐Kovacina J. Integrating Computational and Biological Hemodynamic Approaches to Improve Modeling of Atherosclerotic Arteries. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307627. [PMID: 38704690 PMCID: PMC11234431 DOI: 10.1002/advs.202307627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/12/2024] [Indexed: 05/07/2024]
Abstract
Atherosclerosis is the primary cause of cardiovascular disease, resulting in mortality, elevated healthcare costs, diminished productivity, and reduced quality of life for individuals and their communities. This is exacerbated by the limited understanding of its underlying causes and limitations in current therapeutic interventions, highlighting the need for sophisticated models of atherosclerosis. This review critically evaluates the computational and biological models of atherosclerosis, focusing on the study of hemodynamics in atherosclerotic coronary arteries. Computational models account for the geometrical complexities and hemodynamics of the blood vessels and stenoses, but they fail to capture the complex biological processes involved in atherosclerosis. Different in vitro and in vivo biological models can capture aspects of the biological complexity of healthy and stenosed vessels, but rarely mimic the human anatomy and physiological hemodynamics, and require significantly more time, cost, and resources. Therefore, emerging strategies are examined that integrate computational and biological models, and the potential of advances in imaging, biofabrication, and machine learning is explored in developing more effective models of atherosclerosis.
Collapse
Affiliation(s)
| | - Michael Bartolf‐Kopp
- Department of Functional Materials in Medicine and DentistryInstitute of Functional Materials and Biofabrication (IFB)KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)University of WürzburgPleicherwall 297070WürzburgGermany
| | - Kristina Andelovic
- Department of Functional Materials in Medicine and DentistryInstitute of Functional Materials and Biofabrication (IFB)KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)University of WürzburgPleicherwall 297070WürzburgGermany
| | - Tomasz Jungst
- Department of Functional Materials in Medicine and DentistryInstitute of Functional Materials and Biofabrication (IFB)KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)University of WürzburgPleicherwall 297070WürzburgGermany
- Department of Orthopedics, Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht3584Netherlands
| | - Nona Farbehi
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydney2052Australia
- Tyree Institute of Health EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Garvan Weizmann Center for Cellular GenomicsGarvan Institute of Medical ResearchSydneyNSW2010Australia
| | - Steven G. Wise
- School of Medical SciencesUniversity of SydneySydneyNSW2006Australia
| | - Christopher Hayward
- St Vincent's HospitalSydneyVictor Chang Cardiac Research InstituteSydney2010Australia
| | | | - Jelena Rnjak‐Kovacina
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydney2052Australia
- Tyree Institute of Health EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicine (ACN)University of New South WalesSydneyNSW2052Australia
| |
Collapse
|
5
|
Teuwen JTJ, van der Vorst EPC, Maas SL. Navigating the Maze of Kinases: CaMK-like Family Protein Kinases and Their Role in Atherosclerosis. Int J Mol Sci 2024; 25:6213. [PMID: 38892400 PMCID: PMC11172518 DOI: 10.3390/ijms25116213] [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: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Circulating low-density lipoprotein (LDL) levels are a major risk factor for cardiovascular diseases (CVD), and even though current treatment strategies focusing on lowering lipid levels are effective, CVD remains the primary cause of death worldwide. Atherosclerosis is the major cause of CVD and is a chronic inflammatory condition in which various cell types and protein kinases play a crucial role. However, the underlying mechanisms of atherosclerosis are not entirely understood yet. Notably, protein kinases are highly druggable targets and represent, therefore, a novel way to target atherosclerosis. In this review, the potential role of the calcium/calmodulin-dependent protein kinase-like (CaMKL) family and its role in atherosclerosis will be discussed. This family consists of 12 subfamilies, among which are the well-described and conserved liver kinase B1 (LKB1) and 5' adenosine monophosphate-activated protein kinase (AMPK) subfamilies. Interestingly, LKB1 plays a key role and is considered a master kinase within the CaMKL family. It has been shown that LKB1 signaling leads to atheroprotective effects, while, for example, members of the microtubule affinity-regulating kinase (MARK) subfamily have been described to aggravate atherosclerosis development. These observations highlight the importance of studying kinases and their signaling pathways in atherosclerosis, bringing us a step closer to unraveling the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Jules T. J. Teuwen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80336 München, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
6
|
Lee CH, Lee SH, Kwak HS, Kwak YG, Rosenson RS, Cho YI, Jeong SK. Validation of Signal Intensity Gradient from TOF-MRA for Wall Shear Stress by Phase-Contrast MR. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1248-1258. [PMID: 38332403 PMCID: PMC11169296 DOI: 10.1007/s10278-024-00991-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 02/10/2024]
Abstract
To validate the correlation between the signal intensity gradient (SIG) from time-of-flight magnetic resonance angiography (TOF-MRA) and wall shear stress (WSS) determined by phase contrast magnetic resonance (PC-MR), we conducted both experimental and human studies. In the experimental study, we measured WSS in four tubes of different sizes with variable flow rates using PC-MR and TOF-MRA. The flow rates of water in the experimental study ranged from 0.06 to 12.75 mL/s, resulting in PC-WSS values between 0.1 and 1.6 dyne/cm2. The correlation between PC-WSS and SIG was statistically significant, showing a coefficient of 0.86 (P < 0.001, R2 = 0.75). The line fit provided the conversion equation as Y = 1.6287X - 1.1563 (Y = PC-WSS, X = SIG). For the human study, 28 subjects underwent TOF-MRA and PC-MR examinations of carotid and vertebral arteries. Arterial PC-WSS and SIG were determined in the same segment for each subject. The arterial PC-WSS ranged from 1.9 to 21.0 dyne/cm2. Both carotid and vertebral arteries showed significant correlations between PC-WSS and SIG, with coefficients of 0.85, 0.86, 0.91, and 0.81 in the right and left carotid and vertebral arteries, respectively. Our results show that SIG from TOF-MRA and SIG-WSS derived from the conversion equation provide concurrent in vivo hemodynamic information on arterial shear stress. This study was registered on ClinicalTrials.gov with the identifier NCT04585971 on October 14, 2020.
Collapse
Affiliation(s)
- Chan-Hyuk Lee
- Department of Neurology, Asan Medical Center, Seoul, Republic of Korea
- Department of Neurology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Sang Hyuk Lee
- Equipment Qualification Center for Nuclear Power Plants, Korea Institute of Machinery and Materials, Daejeon, Republic of Korea
| | - Hyo-Sung Kwak
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Yeong-Gon Kwak
- Department of Radiotechnology, Wonkwang Health Science University, Iksan, Republic of Korea
| | - Robert S Rosenson
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Young I Cho
- Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, PA, USA
| | - Seul-Ki Jeong
- Seul-Ki Jeong Neurology Clinic, 233, Gucheonmyeon-ro, Gangdong-gu, Seoul, 05326, Republic of Korea.
| |
Collapse
|
7
|
Tamargo IA, Baek KI, Xu C, Kang DW, Kim Y, Andueza A, Williams D, Demos C, Villa-Roel N, Kumar S, Park C, Choi R, Johnson J, Chang S, Kim P, Tan S, Jeong K, Tsuji S, Jo H. HEG1 Protects Against Atherosclerosis by Regulating Stable Flow-Induced KLF2/4 Expression in Endothelial Cells. Circulation 2024; 149:1183-1201. [PMID: 38099436 PMCID: PMC11001532 DOI: 10.1161/circulationaha.123.064735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/08/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Atherosclerosis preferentially occurs in arterial regions of disturbed blood flow, and stable flow (s-flow) protects against atherosclerosis by incompletely understood mechanisms. METHODS Our single-cell RNA-sequencing data using the mouse partial carotid ligation model was reanalyzed, which identified Heart-of-glass 1 (HEG1) as an s-flow-induced gene. HEG1 expression was studied by immunostaining, quantitive polymerase chain reaction, hybridization chain reaction, and Western blot in mouse arteries, human aortic endothelial cells (HAECs), and human coronary arteries. A small interfering RNA-mediated knockdown of HEG1 was used to study its function and signaling mechanisms in HAECs under various flow conditions using a cone-and-plate shear device. We generated endothelial-targeted, tamoxifen-inducible HEG1 knockout (HEG1iECKO) mice. To determine the role of HEG1 in atherosclerosis, HEG1iECKO and littermate-control mice were injected with an adeno-associated virus-PCSK9 [proprotein convertase subtilisin/kexin type 9] and fed a Western diet to induce hypercholesterolemia either for 2 weeks with partial carotid ligation or 2 months without the surgery. RESULTS S-flow induced HEG1 expression at the mRNA and protein levels in vivo and in vitro. S-flow stimulated HEG1 protein translocation to the downstream side of HAECs and release into the media, followed by increased messenger RNA and protein expression. HEG1 knockdown prevented s-flow-induced endothelial responses, including monocyte adhesion, permeability, and migration. Mechanistically, HEG1 knockdown prevented s-flow-induced KLF2/4 (Kruppel-like factor 2/4) expression by regulating its intracellular binding partner KRIT1 (Krev interaction trapped protein 1) and the MEKK3-MEK5-ERK5-MEF2 pathway in HAECs. Compared with littermate controls, HEG1iECKO mice exposed to hypercholesterolemia for 2 weeks and partial carotid ligation developed advanced atherosclerotic plaques, featuring increased necrotic core area, thin-capped fibroatheroma, inflammation, and intraplaque hemorrhage. In a conventional Western diet model for 2 months, HEG1iECKO mice also showed an exacerbated atherosclerosis development in the arterial tree in both sexes and the aortic sinus in males but not in females. Moreover, endothelial HEG1 expression was reduced in human coronary arteries with advanced atherosclerotic plaques. CONCLUSIONS Our findings indicate that HEG1 is a novel mediator of atheroprotective endothelial responses to flow and a potential therapeutic target.
Collapse
Affiliation(s)
- Ian A. Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, United States
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Chenbo Xu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Dong Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Aitor Andueza
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Darian Williams
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, United States
| | - Catherine Demos
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Nicolas Villa-Roel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Rachel Choi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Janie Johnson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Seowon Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Paul Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Sheryl Tan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Kiyoung Jeong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Shoutaro Tsuji
- Medical Technology & Clinical Engineering, Gunma University of Health and Welfare, Maebashi, Japan
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, United States
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
8
|
Lin A, Ramaswamy Y, Misra A. Developmental heterogeneity of vascular cells: Insights into cellular plasticity in atherosclerosis? Semin Cell Dev Biol 2024; 155:3-15. [PMID: 37316416 DOI: 10.1016/j.semcdb.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Smooth muscle cells, endothelial cells and macrophages display remarkable heterogeneity within the healthy vasculature and under pathological conditions. During development, these cells arise from numerous embryological origins, which confound with different microenvironments to generate postnatal vascular cell diversity. In the atherosclerotic plaque milieu, all these cell types exhibit astonishing plasticity, generating a variety of plaque burdening or plaque stabilizing phenotypes. And yet how developmental origin influences intraplaque cell plasticity remains largely unexplored despite evidence suggesting this may be the case. Uncovering the diversity and plasticity of vascular cells is being revolutionized by unbiased single cell whole transcriptome analysis techniques that will likely continue to pave the way for therapeutic research. Cellular plasticity is only just emerging as a target for future therapeutics, and uncovering how intraplaque plasticity differs across vascular beds may provide key insights into why different plaques behave differently and may confer different risks of subsequent cardiovascular events.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, NSW, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, NSW, Australia; Heart Research Institute, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Baaten CCFMJ, Nagy M, Bergmeier W, Spronk HMH, van der Meijden PEJ. Platelet biology and function: plaque erosion vs. rupture. Eur Heart J 2024; 45:18-31. [PMID: 37940193 PMCID: PMC10757869 DOI: 10.1093/eurheartj/ehad720] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/20/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
The leading cause of heart disease in developed countries is coronary atherosclerosis, which is not simply a result of ageing but a chronic inflammatory process that can lead to acute clinical events upon atherosclerotic plaque rupture or erosion and arterial thrombus formation. The composition and location of atherosclerotic plaques determine the phenotype of the lesion and whether it is more likely to rupture or to erode. Although plaque rupture and erosion both initiate platelet activation on the exposed vascular surface, the contribution of platelets to thrombus formation differs between the two phenotypes. In this review, plaque phenotype is discussed in relation to thrombus composition, and an overview of important mediators (haemodynamics, matrix components, and soluble factors) in plaque-induced platelet activation is given. As thrombus formation on disrupted plaques does not necessarily result in complete vessel occlusion, plaque healing can occur. Therefore, the latest findings on plaque healing and the potential role of platelets in this process are summarized. Finally, the clinical need for more effective antithrombotic agents is highlighted.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, School of Medicine, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
- Blood Research Center, School of Medicine, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Henri M H Spronk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- Thrombosis Expertise Center, Heart+ Vascular Center, Maastricht University Medical Center+, P. Debeyelaan 25, Maastricht, the Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Thrombosis Expertise Center, Heart+ Vascular Center, Maastricht University Medical Center+, P. Debeyelaan 25, Maastricht, the Netherlands
| |
Collapse
|
10
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
11
|
Belhoul-Fakir H, Brown ML, Thompson PL, Hamzah J, Jansen S. Connecting the Dots: How Injury in the Arterial Wall Contributes to Atherosclerotic Disease. Clin Ther 2023; 45:1092-1098. [PMID: 37891144 DOI: 10.1016/j.clinthera.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE The occurrence and development of atherosclerotic cardiovascular disease, which can result in severe outcomes, such as myocardial infarction, stroke, loss of limb, renal failure, and infarction of the gut, are strongly associated with injury to the intimal component of the arterial wall whether via the inside-out or outside-in pathways. The role of injury to the tunica media as a pathway of atherosclerosis initiation is an underresearched area. This review focuses on potential pathways to vessel wall injury as well as current experimental and clinical research in the middle-aged and elderly populations, including the role of exercise, as it relates to injury to the tunica media. METHODS A database search using PubMed and Google Scholar was conducted for research articles published between 1909 and 2023 that focused on pathways of atherogenesis and the impact of mechanical forces on wall injury. The following key words were searched: wall injury, tunica media, atherogenesis, vascular aging, and wall strain. Studies were analyzed, and the relevant information was extracted from each study. FINDINGS A link between high mechanical stress in the arterial wall and reduced vascular compliance was found. The stiffening and calcification of the arterial wall with aging induce high blood pressure and pulse pressure, thereby causing incident hypertension and cardiovascular disease. In turn, prolonged high mechanical stress, particularly wall strain, applied to the arterial wall during vigorous exercise, results in stiffening and calcification of tunica media, accelerated arterial aging, and cardiovascular disease events. In both scenarios, the tunica media is the primary target of mechanical stress and the first to respond to hemodynamic changes. The cyclical nature of these impacts confounds the results of each because they are not mutually exclusive. IMPLICATIONS The role of stress in the tunica media appears to be overlooked despite its relevance, and further research into new primary preventive therapies is needed aside from cautioning the role of vigorous exercise in the elderly population.
Collapse
Affiliation(s)
- Hanane Belhoul-Fakir
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia.
| | - Michael Lawrence Brown
- School of Population Health, Curtin University, Bently, Perth, Western Australia, Australia
| | - Peter L Thompson
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Juliana Hamzah
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Shirley Jansen
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Nedlands, Perth, Western Australia, Australia.
| |
Collapse
|
12
|
Hutton M, Frazer M, Lin A, Patel S, Misra A. New Targets in Atherosclerosis: Vascular Smooth Muscle Cell Plasticity and Macrophage Polarity. Clin Ther 2023; 45:1047-1054. [PMID: 37709601 DOI: 10.1016/j.clinthera.2023.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Despite an increase in treatment options, and substantial reductions in cardiovascular mortality over the past half-century, atherosclerosis remains the most prevalent cause of premature mortality worldwide. The development of innovative new therapies is crucial to further minimize atherosclerosis-related deaths. The diverse array of cell phenotypes derived from vascular smooth muscle cells (SMCs) and macrophages within atherosclerotic plaques are increasingly becoming recognized for their beneficial and detrimental roles in plaque stability and disease burden. This review explores how contemporary transcriptomics and fate-mapping studies have revealed vascular cell plasticity as a relatively unexplored target for therapeutic intervention. METHODS Recent literature for this narrative review was obtained by searching electronic databases (ie, Google Scholar, PubMed). Additional studies were sourced from reference lists and the authors' personal databases. FINDINGS The lipid-rich and inflammatory plaque milieu induces SMC phenotypic switching to both beneficial and detrimental phenotypes. Likewise, macrophage heterogeneity increases with disease burden to a variety of pro-inflammatory and anti-inflammatory activation states. These vascular cell phenotypes are determinants of plaque structure stability, and it is therefore highly likely that they influence clinical outcomes. Development of clinical treatments targeting deleterious phenotypes or promoting pro-healing phenotypes remains in its infancy. However, existing treatments (statins) have shown beneficial effects toward macrophage polarization, providing a rationale for more targeted approaches. In contrast, beneficial SMC phenotypic modulation with these pharmacologic agents has yet to be achieved. The range of modulated vascular cell phenotypes provides a multitude of novel targets and the potential to reduce future adverse events. IMPLICATIONS Vascular cell phenotypic heterogeneity must continue to be explored to lower cardiovascular events in the future. The rapidly increasing weight of evidence surrounding the role of SMC plasticity and macrophage polarity in plaque vulnerability provides a strong foundation upon which development of new therapeutics must follow. This approach may prove to be crucial in reducing cardiovascular events and improving patient benefit in the future.
Collapse
Affiliation(s)
- Michael Hutton
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Madeleine Frazer
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
13
|
Li W, Lin A, Hutton M, Dhaliwal H, Nadel J, Rodor J, Tumanov S, Örd T, Hadden M, Mokry M, Mol BM, Pasterkamp G, Padula MP, Geczy CL, Ramaswamy Y, Sluimer JC, Kaikkonen MU, Stocker R, Baker AH, Fisher EA, Patel S, Misra A. Colchicine promotes atherosclerotic plaque stability independently of inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560632. [PMID: 37873248 PMCID: PMC10592948 DOI: 10.1101/2023.10.03.560632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease which is driven in part by the aberrant trans -differentiation of vascular smooth muscle cells (SMCs). No therapeutic drug has been shown to reverse detrimental SMC-derived cell phenotypes into protective phenotypes, a hypothesized enabler of plaque regression and improved patient outcome. Herein, we describe a novel function of colchicine in the beneficial modulation of SMC-derived cell phenotype, independent of its conventional anti-inflammatory effects. Using SMC fate mapping in an advanced atherosclerotic lesion model, colchicine induced plaque regression by converting pathogenic SMC-derived macrophage-like and osteoblast-like cells into protective myofibroblast-like cells which thickened, and thereby stabilized, the fibrous cap. This was dependent on Notch3 signaling in SMC-derived plaque cells. These findings may help explain the success of colchicine in clinical trials relative to other anti-inflammatory drugs. Thus, we demonstrate the potential of regulating SMC phenotype in advanced plaque regression through Notch3 signaling, in addition to the canonical anti-inflammatory actions of drugs to treat atherosclerosis.
Collapse
|
14
|
Zuin M, Chatzizisis YS, Beier S, Shen C, Colombo A, Rigatelli G. Role of secondary flows in coronary artery bifurcations before and after stenting: What is known so far? CARDIOVASCULAR REVASCULARIZATION MEDICINE 2023; 55:83-87. [PMID: 37385893 DOI: 10.1016/j.carrev.2023.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
Coronary arteries are uniformly exposed to traditional cardiovascular risk factors. However, atherosclerotic lesions occur in preferential regions of the coronary tree, especially in areas with disturbed local blood flow, such as coronary bifurcations. Over the latest years, secondary flows have been linked to the inception and progression of atherosclerosis. Most of these novel findings have been obtained in the field of computational fluid dynamic (CFD) analysis and biomechanics but remain poorly understood by cardiovascular interventionalists, despite the important impact that they may have in clinical practice. We aimed to summarize the current available data regarding the pathophysiological role of secondary flows in coronary artery bifurcation, providing an interpretation of these findings from an interventional perspective.
Collapse
Affiliation(s)
- Marco Zuin
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy.
| | | | - Susann Beier
- School of Mechanical and Manufacturing Engineering, UNSW, Sydney, NSW 2053, Australia
| | - Chi Shen
- School of Mechanical and Manufacturing Engineering, UNSW, Sydney, NSW 2053, Australia
| | - Andrea Colombo
- School of Mechanical and Manufacturing Engineering, UNSW, Sydney, NSW 2053, Australia
| | - Gianluca Rigatelli
- Interventional Cardiology, Department of Cardiology, Aulss6 Euganea, Ospedali Riuniti Padova Sud, Monselice, Italy
| |
Collapse
|
15
|
Brener SJ. How To, Not When To: Treating the Diseased Branch of a Bifurcation Lesion. JACC Cardiovasc Interv 2023; 16:2094-2096. [PMID: 37565965 DOI: 10.1016/j.jcin.2023.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 08/12/2023]
Affiliation(s)
- Sorin J Brener
- Division of Cardiology, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, New York, USA.
| |
Collapse
|
16
|
McLaughlin DK, Hoffmann C, Sasaki M, Li F, Ma J, Cui X, Sutliff RL, Brewster LP. Comparison of arterial storage conditions for delayed arterial ring testing. JVS Vasc Sci 2023; 4:100122. [PMID: 37649473 PMCID: PMC10463248 DOI: 10.1016/j.jvssci.2023.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023] Open
Abstract
Objective Arterial ring testing is the gold standard for measuring arterial function. Increased arterial tone through arterial contraction and impaired endothelial relaxation (endothelial dysfunction) are key metrics of impaired arterial health in peripheral arterial disease (PAD). To allow for comparative testing of arteries during standard laboratory hours, storage buffers and conditions have been used to extend the functional life of arteries. Various storage conditions have been compared, but there has not been a robust comparison or validation in human arteries. The objective of this work is to optimize storage of arterial segments for endothelial cell (EC) testing in a murine model and to test EC function in human PAD arteries. We hypothesized that certain storage conditions would be superior to others. Methods Healthy murine aortas were harvested from 10- to 14-week-old C57/Bl6J male and female mice and compared under different storage protocols (24 hours) to immediate arterial testing. The storage conditions tested were: Opti-MEM (37°C or 4°C), Krebs-HEPES with 1.8 mmol/L or 2.5 mmol/L calcium (4°C), or Wisconsin (WI) solution at 4°C. Vascular function was evaluated by isometric force testing. Endothelium-dependent and -independent relaxation were measured after precontraction with addition of methacholine or sodium nitroprusside, respectively. Arterial contraction was stimulated with potassium chloride or phenylephrine. Analysis of variance was used to determine significance compared with immediate testing with P < .05. Under institutional review board approval, 28 PAD arteries were collected at amputation and underwent vascular function testing as described. Disturbed flow conditions were determined by indirect (upstream occlusion) flow to the harvested tibial arteries. Stable flow arteries had in-line flow. Arterial calcification was quantified manually as present or not present. Results We found that 4°C WI and 37°C Opti-MEM best preserved endothelium-dependent relaxation and performed similarly to immediately testing aortas (termed fresh for freshly tested) (P > .95). Other storage conditions were inferior to freshly tested aortas (P < .05). Vascular smooth muscle function was tested by endothelial-independent relaxation and contractility. All storage conditions preserved endothelial-independent relaxation and contractility similar to freshly tested arteries. However, 4°C WI and 37°C Opti-MEM storage conditions most closely approximated the maximum force of contraction of freshly tested arteries in response to potassium chloride (P > .39). For human arterial testing, 28 tibial arteries were tested for relaxation and contraction with 16 arteries with peripheral artery occlusive disease (PAD with disturbed flow) and 12 without peripheral artery occlusive disease (PAD with stable flow), of which 14 were calcified and 14 were noncalcified. Endothelial-dependent relaxation data was measurable in 9 arteries and arterial contraction data was measurable in 14 arteries. When comparing flow conditions, arteries exposed to disturbed flow (n = 4) had significantly less relaxation (2% vs 59%; P = .03) compared with stable flow conditions (n = 5). In contrast, presence the (n = 6) or absence of calcification (n = 3) did not impact arterial relaxation. Arterial contraction was not different between groups in either comparison by flow (n = 9 disturbed; n = 5 stable) or calcification (n = 6 present; n = 8 absent). Conclusions In healthy murine aortas, arterial storage for 24 hours in 4°C WI or 37°C Opti-MEM both preserved endothelium-dependent relaxation and maximum force of contraction. In human PAD arteries stored in 4° WI, flow conditions before arterial harvest, but not arterial calcification, led to differences in arterial relaxation in human PAD arteries. Arterial contractility was more robust (11/28 arteries) compared with arterial relaxation (7/28 arteries), but was not significantly different under flow or calcification parameters. This work defines ideal storage conditions for arterial ring testing and identifies that EC dysfunction from disturbed flow may persist in delayed ex vivo arterial testing.
Collapse
Affiliation(s)
- Dylan K. McLaughlin
- Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA
- Surgical and Research Services, Atlanta VA Medical Center, Decatur, GA
| | - Carson Hoffmann
- Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA
- Surgical and Research Services, Atlanta VA Medical Center, Decatur, GA
| | - Maiko Sasaki
- Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA
- Surgical and Research Services, Atlanta VA Medical Center, Decatur, GA
| | - Feifei Li
- Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA
- Surgical and Research Services, Atlanta VA Medical Center, Decatur, GA
| | - Jing Ma
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Xiangqin Cui
- Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Roy L. Sutliff
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Luke P. Brewster
- Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA
- Surgical and Research Services, Atlanta VA Medical Center, Decatur, GA
| |
Collapse
|
17
|
Dolmaci OB, Klautz RJM, Poelmann RE, Lindeman JHN, Sprengers R, Kroft L, Grewal N. Thoracic aortic atherosclerosis in patients with a bicuspid aortic valve; a case-control study. BMC Cardiovasc Disord 2023; 23:363. [PMID: 37468858 DOI: 10.1186/s12872-023-03396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/13/2023] [Indexed: 07/21/2023] Open
Abstract
INTRODUCTION Bicuspid aortic valve (BAV) patients have an increased risk to develop thoracic aortic complications. Little is known about the prevalence and severity of atherosclerosis in the BAV ascending aortic wall. This study evaluates and compares the prevalence of thoracic aortic atherosclerosis in BAV and tricuspid aortic valve (TAV) patients. METHODS Atherosclerosis was objectified using three diagnostic modalities in two separate BAV patient cohorts (with and without an aortic dilatation). Within the first group, atherosclerosis was graded histopathologically according to the modified AHA classification scheme proposed by Virmani et al. In the second group, the calcific load of the ascending aorta and coronary arteries, coronary angiographies and cardiovascular risk factors were studied. Patients were selected from a surgical database (treated between 2006-2020), resulting in a total of 128 inclusions. RESULTS Histopathology showed atherosclerotic lesions to be more prevalent and severe in all TAV as compared to all BAV patients (OR 1.49 (95%CI 1.14 - 1.94); p = 0.003). Computed tomography showed no significant differences in ascending aortic wall calcification between all BAV and all TAV patients, although a tendency of lower calcific load in favor of BAV was seen. Coronary calcification was higher in all TAV as compared to all BAV (OR 1.30 (95%CI 1.06 - 1.61); p = 0.014). CONCLUSION Ascending aortic atherosclerotic plaques were histologically more pronounced in TAV as compared to the BAV patients, while CT scans revealed equal amounts of calcific depositions within the ascending aortic wall. This study confirms less atherosclerosis in the ascending aortic wall and coronary arteries of BAV patients as compared to TAV patients. These results were not affected by the presence of a thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Onur B Dolmaci
- Department of Cardiothoracic Surgery, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Robert J M Klautz
- Department of Cardiothoracic Surgery, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Robert E Poelmann
- Institute of Biology, Animal Sciences and Health, Leiden University, Leiden, The Netherlands
| | - Jan H N Lindeman
- Department of Vascular Surgery, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Ralf Sprengers
- Department of Radiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lucia Kroft
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nimrat Grewal
- Department of Cardiothoracic Surgery, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands.
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
18
|
Vizcardo-Galindo GA, Howe CA, Hoiland RL, Carter HH, Willie CK, Ainslie PN, Tremblay JC. Impact of Oxygen Supplementation on Brachial Artery Hemodynamics and Vascular Function During Ascent to 5,050 m. High Alt Med Biol 2023; 24:27-36. [PMID: 36940101 DOI: 10.1089/ham.2022.0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Vizcardo-Galindo, Gustavo A., Connor A. Howe, Ryan L. Hoiland, Howard H. Carter, Christopher K. Willie, Philip N. Ainslie, and Joshua C. Tremblay. Impact of oxygen supplementation on brachial artery hemodynamics and vascular function during ascent to 5,050 m. High Alt Med Biol. 24:27-36, 2023.-High-altitude trekking alters upper limb hemodynamics and reduces brachial artery vascular function in lowlanders. Whether these changes are reversible with the removal of hypoxia is unknown. We investigated the impact of 20 minutes of oxygen supplementation (O2) on brachial artery hemodynamics, reactive hyperemia (RH; microvascular function), and flow-mediated dilation (FMD; endothelial function). Participants (aged 21-42 years) were examined before and with O2 at 3,440 m (n = 7), 4,371 m (n = 7), and 5,050 m (n = 12) using Duplex ultrasound (days 4, 7, and 10 respectively). At 3,440 m, O2 decreased brachial artery diameter (-5% ± 5%; p = 0.04), baseline blood flow (-44% ± 15%; p < 0.001), oxygen delivery (-39 ± 16; p < 0.001), and peak RH (-8% ± 8%; p = 0.02), but not RH normalized for baseline blood flow. Elevated FMD (p = 0.04) with O2 at 3,440 m was attributed to the reduction in baseline diameter. At 5,050 m, a reduction in brachial artery blood flow (-17% ± 22%; p = 0.03), but not oxygen delivery, diameter, RH, or FMD occurred with O2. These findings suggest that during early trekking at high altitude, O2 causes vasoconstriction in the upper limb along the arterial tree (conduit and resistance arteries). With incremental high-altitude exposure, O2 reduces blood flow without compromising oxygen delivery, RH, or FMD, suggesting a differential impact on vascular function modulated by the duration and severity of high-altitude exposure.
Collapse
Affiliation(s)
- Gustavo A Vizcardo-Galindo
- Centre for Heart, Lung & Vascular Health, Faculty of Health and Social Development, University of British Columbia-Okanagan, Kelowna, Canada
| | - Connor A Howe
- Centre for Heart, Lung & Vascular Health, Faculty of Health and Social Development, University of British Columbia-Okanagan, Kelowna, Canada
| | - Ryan L Hoiland
- Centre for Heart, Lung & Vascular Health, Faculty of Health and Social Development, University of British Columbia-Okanagan, Kelowna, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- International Collaboration on Repair Discoveries, Vancouver, Canada
| | - Howard H Carter
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Cardiovascular Research Group, School of Human Sciences (Exercise and Sport Science), University of Western Australia, Perth, Australia
| | - Christopher K Willie
- Centre for Heart, Lung & Vascular Health, Faculty of Health and Social Development, University of British Columbia-Okanagan, Kelowna, Canada
| | - Philip N Ainslie
- Centre for Heart, Lung & Vascular Health, Faculty of Health and Social Development, University of British Columbia-Okanagan, Kelowna, Canada
| | - Joshua C Tremblay
- Centre for Heart, Lung & Vascular Health, Faculty of Health and Social Development, University of British Columbia-Okanagan, Kelowna, Canada
| |
Collapse
|
19
|
Çap M, Torii R, Onuma Y, Krams R, Bennett MR, Stone PH, Serruys PW, Bourantas CV. Editorial: Computational modeling for assessing coronary artery pathophysiology. Front Cardiovasc Med 2023; 10:1113835. [PMID: 36733302 PMCID: PMC9887330 DOI: 10.3389/fcvm.2023.1113835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Affiliation(s)
- Murat Çap
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Department of Cardiology, University of Health Sciences Diyarbakir Gazi Yaşargil Education and Research Hospital, Diyarbakir, Turkey
| | - Ryo Torii
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Yoshinobu Onuma
- Department of Cardiology, National University of Ireland Galway (NUIG), Galway, Ireland
| | - Rob Krams
- Department of Molecular Bioengineering Engineering and Material Sciences, Queen Mary University of London, London, United Kingdom
| | - Martin R. Bennett
- Division of Cardiovascular Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Peter H. Stone
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Patrick W. Serruys
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Christos V. Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
20
|
Tauchi M, Oshita K, Urschel K, Furtmair R, Kühn C, Stumpfe FM, Botos B, Achenbach S, Dietel B. The Involvement of Cx43 in JNK1/2-Mediated Endothelial Mechanotransduction and Human Plaque Progression. Int J Mol Sci 2023; 24:ijms24021174. [PMID: 36674690 PMCID: PMC9863493 DOI: 10.3390/ijms24021174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023] Open
Abstract
Atherosclerotic lesions preferentially develop at bifurcations, characterized by non-uniform shear stress (SS). The aim of this study was to investigate SS-induced endothelial activation, focusing on stress-regulated mitogen-activated protein kinases (MAPK) and downstream signaling, and its relation to gap junction proteins, Connexins (Cxs). Human umbilical vein endothelial cells were exposed to flow ("mechanical stimulation") and stimulated with TNF-α ("inflammatory stimulation"). Phosphorylated levels of MAPKs (c-Jun N-terminal kinase (JNK1/2), extracellular signal-regulated kinase (ERK), and p38 kinase (p38K)) were quantified by flow cytometry, showing the activation of JNK1/2 and ERK. THP-1 cell adhesion under non-uniform SS was suppressed by the inhibition of JNK1/2, not of ERK. Immunofluorescence staining and quantitative real-time PCR demonstrated an induction of c-Jun and c-Fos and of Cx43 in endothelial cells by non-uniform SS, and the latter was abolished by JNK1/2 inhibition. Furthermore, plaque inflammation was analyzed in human carotid plaques (n = 40) using immunohistochemistry and quanti-gene RNA-assays, revealing elevated Cx43+ cell counts in vulnerable compared to stable plaques. Cx43+ cell burden in the plaque shoulder correlated with intraplaque neovascularization and lipid core size, while an inverse correlation was observed with fibrous cap thickness. Our results constitute the first report that JNK1/2 mediates Cx43 mechanoinduction in endothelial cells by atheroprone shear stress and that Cx43 is expressed in human carotid plaques. The correlation of Cx43+ cell counts with markers of plaque vulnerability implies its contribution to plaque progression.
Collapse
Affiliation(s)
- Miyuki Tauchi
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Cognitive and Molecular Research Institute of Brain Diseases, Kurume University, Kurume 830-0011, Japan
| | - Kensuke Oshita
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Department of Anesthesiology, School of Medicine, Kurume University, Kurume 830-0011, Japan
| | - Katharina Urschel
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Roman Furtmair
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Constanze Kühn
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Florian M. Stumpfe
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Balazs Botos
- Department of Vascular Surgery, Hospital of Nürnberg-Süd, 90471 Nürnberg, Germany
| | - Stephan Achenbach
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Barbara Dietel
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Correspondence:
| |
Collapse
|
21
|
Hurd ER, Iffrig E, Jiang D, Oshinski JN, Timmins LH. Flow-based method demonstrates improved accuracy for calculating wall shear stress in arterial flows from 4D flow MRI data. J Biomech 2023; 146:111413. [PMID: 36535100 PMCID: PMC9845191 DOI: 10.1016/j.jbiomech.2022.111413] [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: 06/15/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Four-dimensional flow magnetic resonance imaging (i.e., 4D flow MRI) has become a valuable tool for the in vivo assessment of blood flow within large vessels and cardiac chambers. As wall shear stress (WSS) has been correlated with the development and progression of cardiovascular disease, focus has been directed at developing techniques to quantify WSS directly from 4D flow MRI data. The goal of this study was to compare the accuracy of two such techniques - termed the velocity and flow-based methods - in the setting of simplified and complex flow scenarios. Synthetic MR data were created from exact solutions to the Navier-Stokes equations for the steady and pulsatile flow of an incompressible, Newtonian fluid through a rigid cylinder. In addition, synthetic MR data were created from the predicted velocity fields derived from a fluid-structure interaction (FSI) model of pulsatile flow through a thick-walled, multi-layered model of the carotid bifurcation. Compared to the analytical solutions for steady and pulsatile flow, the flow-based method demonstrated greater accuracy than the velocity-based method in calculating WSS across all changes in fluid velocity/flow rate, tube radius, and image signal-to-noise (p < 0.001). Furthermore, the velocity-based method was more sensitive to boundary segmentation than the flow-based method. When compared to results from the FSI model, the flow-based method demonstrated greater accuracy than the velocity-based method with average differences in time-averaged WSS of 0.31 ± 1.03 Pa and 0.45 ± 1.03 Pa, respectively (p <0.005). These results have implications on the utility, accuracy, and clinical translational of methods to determine WSS from 4D flow MRI.
Collapse
Affiliation(s)
- Elliott R Hurd
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Elizabeth Iffrig
- Division of Allergy, Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - David Jiang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - John N Oshinski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lucas H Timmins
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
22
|
Ouyang Z, Zhong J, Shen J, Zeng Y. The cell origins of foam cell and lipid metabolism regulated by mechanical stress in atherosclerosis. Front Physiol 2023; 14:1179828. [PMID: 37123258 PMCID: PMC10133704 DOI: 10.3389/fphys.2023.1179828] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Atherosclerosis is an inflammatory disease initiated by endothelial activation, in which lipoprotein, cholesterol, extracellular matrix, and various types of immune and non-immune cells are accumulated and formed into plaques on the arterial wall suffering from disturbed flow, characterized by low and oscillating shear stress. Foam cells are a major cellular component in atherosclerotic plaques, which play an indispensable role in the occurrence, development and rupture of atherosclerotic plaques. It was previously believed that foam cells were derived from macrophages or smooth muscle cells, but recent studies have suggested that there are other sources of foam cells. Many studies have found that the distribution of atherosclerotic plaques is not random but distributed at the bend and bifurcation of the arterial tree. The development and rupture of atherosclerotic plaque are affected by mechanical stress. In this review, we reviewed the advances in foam cell formation in atherosclerosis and the regulation of atherosclerotic plaque and lipid metabolism by mechanical forces. These findings provide new clues for investigating the mechanisms of atherosclerotic plaque formation and progression.
Collapse
|
23
|
Schultz J, van den Hoogen IJ, Kuneman JH, de Graaf MA, Kamperidis V, Broersen A, Jukema JW, Sakellarios A, Nikopoulos S, Tsarapatsani K, Naka K, Michalis L, Fotiadis DI, Maaniitty T, Saraste A, Bax JJ, Knuuti J. Coronary computed tomography angiography-based endothelial wall shear stress in normal coronary arteries. Int J Cardiovasc Imaging 2023; 39:441-450. [PMID: 36255544 PMCID: PMC9870961 DOI: 10.1007/s10554-022-02739-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/27/2022] [Indexed: 01/27/2023]
Abstract
Endothelial wall shear stress (ESS) is a biomechanical force which plays a role in the formation and evolution of atherosclerotic lesions. The purpose of this study is to evaluate coronary computed tomography angiography (CCTA)-based ESS in coronary arteries without atherosclerosis, and to assess factors affecting ESS values. CCTA images from patients with suspected coronary artery disease were analyzed to identify coronary arteries without atherosclerosis. Minimal and maximal ESS values were calculated for 3-mm segments. Factors potentially affecting ESS values were examined, including sex, lumen diameter and distance from the ostium. Segments were categorized according to lumen diameter tertiles into small (< 2.6 mm), intermediate (2.6-3.2 mm) or large (≥ 3.2 mm) segments. A total of 349 normal vessels from 168 patients (mean age 59 ± 9 years, 39% men) were included. ESS was highest in the left anterior descending artery compared to the left circumflex artery and right coronary artery (minimal ESS 2.3 Pa vs. 1.9 Pa vs. 1.6 Pa, p < 0.001 and maximal ESS 3.7 Pa vs. 3.0 Pa vs. 2.5 Pa, p < 0.001). Men had lower ESS values than women, also after adjusting for lumen diameter (p < 0.001). ESS values were highest in small segments compared to intermediate or large segments (minimal ESS 3.8 Pa vs. 1.7 Pa vs. 1.2 Pa, p < 0.001 and maximal ESS 6.0 Pa vs. 2.6 Pa vs. 2.0 Pa, p < 0.001). A weak to strong correlation was found between ESS and distance from the ostium (ρ = 0.22-0.62, p < 0.001). CCTA-based ESS values increase rapidly and become widely scattered with decreasing lumen diameter. This needs to be taken into account when assessing the added value of ESS beyond lumen diameter in highly stenotic lesions.
Collapse
Affiliation(s)
- Jussi Schultz
- grid.410552.70000 0004 0628 215XTurku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| | - Inge J. van den Hoogen
- grid.10419.3d0000000089452978Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jurrien H. Kuneman
- grid.10419.3d0000000089452978Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michiel A. de Graaf
- grid.10419.3d0000000089452978Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vasileios Kamperidis
- Department of Cardiology, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexander Broersen
- grid.10419.3d0000000089452978Department of Radiology, Division of Image Processing, Leiden University Medical Center, Leiden, The Netherlands
| | - J. Wouter Jukema
- grid.10419.3d0000000089452978Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands ,grid.411737.7Netherlands Heart Institute, Utrecht, The Netherlands
| | - Antonis Sakellarios
- Department of Biomedical Research, FORTH-IMBB, Ioannina, Greece ,grid.9594.10000 0001 2108 7481Department of Materials Science and Engineering, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Sotirios Nikopoulos
- grid.9594.10000 0001 2108 7481Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Konstantina Tsarapatsani
- Department of Biomedical Research, FORTH-IMBB, Ioannina, Greece ,grid.9594.10000 0001 2108 7481Department of Materials Science and Engineering, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Katerina Naka
- grid.9594.10000 0001 2108 7481Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Lampros Michalis
- grid.9594.10000 0001 2108 7481Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Dimitrios I. Fotiadis
- Department of Biomedical Research, FORTH-IMBB, Ioannina, Greece ,grid.9594.10000 0001 2108 7481Department of Materials Science and Engineering, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Teemu Maaniitty
- grid.410552.70000 0004 0628 215XTurku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| | - Antti Saraste
- grid.410552.70000 0004 0628 215XTurku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, 20520 Turku, Finland ,grid.410552.70000 0004 0628 215XHeart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Jeroen J. Bax
- grid.10419.3d0000000089452978Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands ,grid.410552.70000 0004 0628 215XHeart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Juhani Knuuti
- grid.410552.70000 0004 0628 215XTurku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, 20520 Turku, Finland ,grid.10419.3d0000000089452978Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Badour MI, Stone RM, Parikh KS, Lester NJ, Meloche OL, Wulterkens RN, Bain AR. Circulating Notch1 in response to altered vascular wall shear stress in adults. Exp Physiol 2022; 107:1426-1431. [PMID: 36116111 DOI: 10.1113/ep090749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? Is the plasma concentration of Notch1 extracellular domain altered in response to decreased and increased vascular wall shear stress in the forearm in humans? What is the main finding and its importance? Notch1 extracellular domain is increased with acute increases in antegrade shear rate but does not change with 20 min of decreased shear rate caused by distal forearm occlusion. A novel and integral endothelial mechanosensor in humans that can help explain vascular endothelial adjustments in response to increases in antegrade shear stress was characterized. ABSTRACT Notch1 has been proposed as a novel endothelial mechanosensor that is central for signalling adjustments in response to changes in vascular wall shear stress. However, there remains no controlled in vivo study in humans. Accordingly, we sought to address the question of whether plasma concentrations of Notch1 extracellular domain (ECD) is altered in response to transient changes in vascular wall shear stress. In 10 young healthy adults (6M/4F), alterations in shear stress were induced by supra-systolic cuff inflation around the wrist. The opposite arm was treated as a time control with no wrist cuff inflation. Plasma was collected from an antecubital vein of both arms at baseline, 20 min of wrist cuff inflation (low shear), as well as 1-2 min (high shear) and 15 min following (recovery) wrist cuff release. The Notch1 ECD was quantified using a commercially available ELISA. Duplex ultrasound was used to confirm alterations in shear stress. In the experimental arm, concentrations of Notch1 ECD remained statistically similar to baseline at all time points except for immediately following cuff release where it was elevated by ∼50% (P = 0.033), coinciding with the condition of high antegrade shear rate. Concentrations of Notch1 ECD remained unchanged in the control arm through all time points. These data indicate that Notch1 is a viable biomarker for quantifying mechanotransduction in response to increased shear stress in humans, and it may underlie the vascular adaptations or mal-adaptations associated with conditions that impact antegrade shear.
Collapse
Affiliation(s)
- Matthew I Badour
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| | - Rachel M Stone
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| | | | | | - Olivia L Meloche
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| | | | - Anthony R Bain
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| |
Collapse
|
25
|
van den Hoogen IJ, Schultz J, Kuneman JH, de Graaf MA, Kamperidis V, Broersen A, Jukema JW, Sakellarios A, Nikopoulos S, Kyriakidis S, Naka KK, Michalis L, Fotiadis DI, Maaniitty T, Saraste A, Bax JJ, Knuuti J. Detailed behaviour of endothelial wall shear stress across coronary lesions from non-invasive imaging with coronary computed tomography angiography. Eur Heart J Cardiovasc Imaging 2022; 23:1708-1716. [PMID: 35616068 PMCID: PMC10017098 DOI: 10.1093/ehjci/jeac095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/15/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Evolving evidence suggests that endothelial wall shear stress (ESS) plays a crucial role in the rupture and progression of coronary plaques by triggering biological signalling pathways. We aimed to investigate the patterns of ESS across coronary lesions from non-invasive imaging with coronary computed tomography angiography (CCTA), and to define plaque-associated ESS values in patients with coronary artery disease (CAD). METHODS AND RESULTS Symptomatic patients with CAD who underwent a clinically indicated CCTA scan were identified. Separate core laboratories performed blinded analysis of CCTA for anatomical and ESS features of coronary atherosclerosis. ESS was assessed using dedicated software, providing minimal and maximal ESS values for each 3 mm segment. Each coronary lesion was divided into upstream, start, minimal luminal area (MLA), end and downstream segments. Also, ESS ratios were calculated using the upstream segment as a reference. From 122 patients (mean age 64 ± 7 years, 57% men), a total of 237 lesions were analyzed. Minimal and maximal ESS values varied across the lesions with the highest values at the MLA segment [minimal ESS 3.97 Pa (IQR 1.93-8.92 Pa) and maximal ESS 5.64 Pa (IQR 3.13-11.21 Pa), respectively]. Furthermore, minimal and maximal ESS values were positively associated with stenosis severity (P < 0.001), percent atheroma volume (P < 0.001), and lesion length (P ≤ 0.023) at the MLA segment. Using ESS ratios, similar associations were observed for stenosis severity and lesion length. CONCLUSIONS Detailed behaviour of ESS across coronary lesions can be derived from routine non-invasive CCTA imaging. This may further improve risk stratification.
Collapse
Affiliation(s)
| | - Jussi Schultz
- Turku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, Turku 20520, Finland
| | - Jurrien H Kuneman
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michiel A de Graaf
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vasileios Kamperidis
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alexander Broersen
- Department of Radiology, Division of Image Processing, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Antonis Sakellarios
- Department of Biomedical Research, FORTH-IMBB, Ioannina, Greece.,Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
| | - Sotirios Nikopoulos
- Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Savvas Kyriakidis
- Department of Biomedical Research, FORTH-IMBB, Ioannina, Greece.,Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Lampros Michalis
- Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Dimitrios I Fotiadis
- Department of Biomedical Research, FORTH-IMBB, Ioannina, Greece.,Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
| | - Teemu Maaniitty
- Turku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, Turku 20520, Finland
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, Turku 20520, Finland.,Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Juhani Knuuti
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Turku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, Turku 20520, Finland
| |
Collapse
|
26
|
Katakia YT, Kanduri S, Bhattacharyya R, Ramanathan S, Nigam I, Kuncharam BVR, Majumder S. Angular difference in human coronary artery governs endothelial cell structure and function. Commun Biol 2022; 5:1044. [PMID: 36183045 PMCID: PMC9526720 DOI: 10.1038/s42003-022-04014-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
Blood vessel branch points exhibiting oscillatory/turbulent flow and lower wall shear stress (WSS) are the primary sites of atherosclerosis development. Vascular endothelial functions are essentially dependent on these tangible biomechanical forces including WSS. Herein, we explored the influence of blood vessel bifurcation angles on hemodynamic alterations and associated changes in endothelial function. We generated computer-aided design of a branched human coronary artery followed by 3D printing such designs with different bifurcation angles. Through computational fluid dynamics analysis, we observed that a larger branching angle generated more complex turbulent/oscillatory hemodynamics to impart minimum WSS at branching points. Through the detection of biochemical markers, we recorded significant alteration in eNOS, ICAM1, and monocyte attachment in EC grown in microchannel having 60o vessel branching angle which correlated with the lower WSS. The present study highlights the importance of blood vessel branching angle as one of the crucial determining factors in governing atherogenic-endothelial dysfunction. In silico and in vitro investigations reveal angular differences in the blood vessel branching points differentially alter the hemodynamics to impact endothelial structure and function.
Collapse
Affiliation(s)
- Yash T Katakia
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Satyadevan Kanduri
- Department of Chemical Engineering, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ritobrata Bhattacharyya
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Srinandini Ramanathan
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ishan Nigam
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | | | - Syamantak Majumder
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India.
| |
Collapse
|
27
|
Feng S, Chen JW, Shu XY, Aihemaiti M, Quan JW, Lu L, Zhang RY, Yang CD, Wang XQ. Endothelial microparticles: A mechanosensitive regulator of vascular homeostasis and injury under shear stress. Front Cell Dev Biol 2022; 10:980112. [PMID: 36172284 PMCID: PMC9510576 DOI: 10.3389/fcell.2022.980112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Hemodynamic shear stress (SS), a frictional force generated by blood flow, regulates vascular homeostasis. High and steady SS maintains physiological function of endothelial cells while low and disturbed SS promotes disturbance of vascular homeostasis and the development of atherosclerosis. Endothelial microparticle (EMP), a vesicular structure shed from endothelial cells, has emerged as a surrogate biomarker of endothelial injury and dysfunction. EMP release is triggered by disturbed SS in addition to multiple inflammatory cytokines. This review systematically summarizes the impact of SS on EMPs and the role of EMPs under SS in modulating vascular homeostasis and injury, including endothelial survival, vasodilation, inflammatory response, vascular permeability, and coagulation system.
Collapse
Affiliation(s)
- Shuo Feng
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Jia Wei Chen
- Institute of Cardiovascular Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Xin Yi Shu
- Institute of Cardiovascular Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Muladili Aihemaiti
- Institute of Cardiovascular Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Jin Wei Quan
- Institute of Cardiovascular Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Rui Yan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Chen Die Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiao Qun Wang, ; Chen Die Yang,
| | - Xiao Qun Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Disease, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiao Qun Wang, ; Chen Die Yang,
| |
Collapse
|
28
|
Wang G, Li Z, Chen C, Yan F, Wei J, Zhang Z, Chen Y. The hemodynamic effect of eccentricity in visceral branched aneurysms with multilayer stents. Proc Inst Mech Eng H 2022; 236:1070-1079. [DOI: 10.1177/09544119221106829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It is preliminarily acknowledged that multilayer stent (MS) is a promising alternative technology in the treatment of visceral branched aneurysms, but hemodynamic consequences of eccentricity in such aneurysms with MS are less examined. In this work, we performed a time-dependent simulation of branched aneurysms of various eccentricities with different stent layers, and thrombosis-related parameters, such as time-averaged wall shear stress (TAWSS), oscillating shear index (OSI), and relative residence time (RRT), were also analyzed. Our results revealed that MS can generally restore laminar flow inside the stent, and allow proper perfusion to vital organs while also fostering a relatively secluded hemodynamic environment for thrombosis formation. Particularly, a flow in the aneurysm sac communicating between the main artery and side branch forms at early systole. However, MS fails to completely eliminate detrimental flow impingement after peak systole, which may hinder aneurysm recovery, especially in the cases of eccentric aneurysms. Therefore, saccular aneurysms should be treated with more caution than fusiform aneurysms. And further therapeutic attempts to keep both perfusion in the proximal region of the aneurysm and isolation in the distal region of the aneurysm should be considered.
Collapse
Affiliation(s)
- Guanshi Wang
- Laboratory of Biomechanical Engineering, Department of Applied Mechanics, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Zhongyou Li
- Laboratory of Biomechanical Engineering, Department of Applied Mechanics, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Chong Chen
- Laboratory of Biomechanical Engineering, Department of Applied Mechanics, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Fei Yan
- Interdisciplinary Division of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Junru Wei
- Laboratory of Biomechanical Engineering, Department of Applied Mechanics, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Zhuo Zhang
- College of Mechanical Engineering, Sichuan University, Chengdu, China
| | - Yu Chen
- Laboratory of Biomechanical Engineering, Department of Applied Mechanics, College of Architecture and Environment, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Gu SZ, Bennett MR. Plaque Structural Stress: Detection, Determinants and Role in Atherosclerotic Plaque Rupture and Progression. Front Cardiovasc Med 2022; 9:875413. [PMID: 35872913 PMCID: PMC9300846 DOI: 10.3389/fcvm.2022.875413] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/10/2022] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis remains a major cause of death worldwide, with most myocardial infarctions being due to rupture or erosion of coronary plaques. Although several imaging modalities can identify features that confer risk, major adverse cardiovascular event (MACE) rates attributable to each plaque are low, such that additional biomarkers are required to improve risk stratification at plaque and patient level. Coronary arteries are exposed to continual mechanical forces, and plaque rupture occurs when plaque structural stress (PSS) exceeds its mechanical strength. Prospective studies have shown that peak PSS is correlated with acute coronary syndrome (ACS) presentation, plaque rupture, and MACE, and provides additional prognostic information to imaging. In addition, PSS incorporates multiple variables, including plaque architecture, plaque material properties, and haemodynamic data into a defined solution, providing a more detailed overview of higher-risk lesions. We review the methods for calculation and determinants of PSS, imaging modalities used for modeling PSS, and idealized models that explore structural and geometric components that affect PSS. We also discuss current experimental and clinical data linking PSS to the natural history of coronary artery disease, and explore potential for refining treatment options and predicting future events.
Collapse
Affiliation(s)
| | - Martin R. Bennett
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Nooraeen A, Ghalichi F, Taghizadeh H, Guidoin R. Probing the possibility of lesion formation/progression in vicinity of a primary atherosclerotic plaque: A fluid-solid interaction study and angiographic evidences. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2022; 38:e3605. [PMID: 35481668 DOI: 10.1002/cnm.3605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
It is shown that certain locations in the arterial tree, such as coronary and cerebral arteries, are more prevalent to plaque formation. Endothelial activation and consequent plaque development are attributed to local hemodynamic parameters such as wall shear stress (WSS), oscillatory shear index (OSI), relative residence time (RRT), and stress phase angle. After a certain level of plaque progression, these hemodynamic parameters are disturbed before and after the plaque. In the current study, it is hypothesized that the vicinity of a primary lesion is susceptible for further degeneration and second plaque formation. A fluid-solid interaction (FSI) model of the coronary artery with different levels of asymmetric constriction, is simulated and the trend of hemodynamic parameters were studied in both of the plaque side (PS) and the opposite wall (facing the plaque [PF]). Also, a novel factor is introduced that can identify the high-risk regions associated with WSS oscillations to negative values. Our results indicate that when more than half of the artery is constricted, the downstream of the plaque is highly exposed to endothelial pathogenesis the PS, such that negative WSS, and as well, critical values of OSI and RRT, that is, -1.2 Pa, 0.42 and 6.5 s, respectively arise in this region. PS endothelial cells in this region exposed to the highest risk of atherosclerosis based on the proposed index (3 out of 3). As well, three cases of angiographic images are provided that confirms existence of secondary lesion close to the primary one as predicted by our computational simulations.
Collapse
Affiliation(s)
- Ahmad Nooraeen
- Tissue Mechanics Lab, Faculty of Biomedical Engineering, Sahand University of Technology, Tabriz, Iran
| | - Farzan Ghalichi
- Faculty of Biomedical Engineering, Sahand University of Technology, Tabriz, Iran
| | - Hadi Taghizadeh
- Tissue Mechanics Lab, Faculty of Biomedical Engineering, Sahand University of Technology, Tabriz, Iran
| | - Robert Guidoin
- Department of Surgery, Université Laval and Division of Regenerative Medicine, CHU de Québec Research Centre, Quebec, Canada
| |
Collapse
|
31
|
Wu J, Wang P, Zhou L, Zhang D, Chen Q, Mao C, Su W, Huo Y, Peng J, Yin X, Chen G. Hemodynamics derived from computational fluid dynamics based on magnetic resonance angiography is associated with functional outcomes in atherosclerotic middle cerebral artery stenosis. Quant Imaging Med Surg 2022; 12:688-698. [PMID: 34993111 DOI: 10.21037/qims-21-337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/23/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND To investigate the relationship between fluid-attenuated inversion recovery (FLAIR) vascular hyperintensity (FVH), hemodynamics, and functional outcome in atherosclerotic middle cerebral artery (MCA) stenosis using a computational fluid dynamics (CFD) model based on magnetic resonance angiography (MRA), according to a modified Rankin Scale (mRS) at 3 months. METHODS A total of 120 patients with 50-99% atherosclerotic MCA stenosis were included. The training and internal validation groups were composed of 99 participants and 21 participants, respectively. Demographic, imaging data, and functional outcome (mRS at 3 months) were collected. Hemodynamic parameters were obtained from the CFD model. The FVH score was based on the number of territories where FVH is positive, according to the spatial distribution in the Alberta Stroke Program Early Computed Tomography Score (ASPECTS). The prediction models were constructed according to clinical and hemodynamic parameters using multivariate logistic analysis. The DeLong test compared areas under the curves (AUCs) of the models. RESULTS The multivariable logistic regression analysis showed that the National Institute of Health Stroke Scale (NIHSS) at admission, hypertension, hyperlipidemia, the ratio of wall shear stress before treatment (WSSRbefore), and difference in the ratio of wall shear stress (WSSR) were independently associated with functional outcome (all P<0.05). In the training group before treatment, the AUC of model 1a (only clinical variables) and 2a (clinical variables with addition of WSSRbefore) were 0.750 and 0.802. After treatment, the AUC of model 1b (only clinical variables) and 2b (clinical variables with addition of difference in WSSR) were 0.815 and 0.883, respectively. The AUC of models with hemodynamic parameters was significantly higher than the models based on clinical variables only (all P<0.05, DeLong test). In the internal validation group before treatment, the AUC of the model (clinical variables) was 0.782, and that of the model (clinical variables and WSSRbefore) was 0.800. After treatment, the AUC of the model (clinical variables) was 0.833, and that of the model (clinical variables and difference in WSSR) was 0.861. There were no significant differences between the good and the poor functional outcome group concerning FVHbefore scores before treatment (0.30±0.81 vs. 0.26±0.97; P=0.321) and FVHafter scores after treatment (0.08±0.39 vs. 0.00±0.00; P=0.244). CONCLUSIONS Hemodynamics was associated with functional outcomes in patients with ischemic stroke attributed to atherosclerotic MCA stenosis, while FVH was not. Hemodynamic parameters were of great importance in the prediction models.
Collapse
Affiliation(s)
- Jiahua Wu
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Wang
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Leilei Zhou
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Danfeng Zhang
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Cunnan Mao
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wen Su
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yingsong Huo
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Peng
- Intervention Department, Chenggong Hospital Affiliated to Xiamen University, Xiamen, China
| | - Xindao Yin
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guozhong Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Adriaenssens T, Allard-Ratick MP, Thondapu V, Sugiyama T, Raffel OC, Barlis P, Poon EKW, Araki M, Nakajima A, Minami Y, Takano M, Kurihara O, Fuster V, Kakuta T, Jang IK. Optical Coherence Tomography of Coronary Plaque Progression and Destabilization: JACC Focus Seminar Part 3/3. J Am Coll Cardiol 2021; 78:1275-1287. [PMID: 34531029 DOI: 10.1016/j.jacc.2021.07.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
The development of optical coherence tomography (OCT) has revolutionized our understanding of coronary artery disease. In vivo OCT research has paralleled with advances in computational fluid dynamics, providing additional insights in the various hemodynamic factors influencing plaque growth and stability. Recent OCT studies introduced a new concept of plaque healing in relation to clinical presentation. In addition to known mechanisms of acute coronary syndromes such as plaque rupture and plaque erosion, a new classification of calcified plaque was recently reported. This review will focus on important new insights that OCT has provided in recent years into coronary plaque development, progression, and destabilization, with a focus on the role of local hemodynamics and endothelial shear stress, the layered plaque (signature of previous subclinical plaque destabilization and healing), and the calcified culprit plaque.
Collapse
Affiliation(s)
- Tom Adriaenssens
- Department of Cardiovascular Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Marc P Allard-Ratick
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vikas Thondapu
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomoyo Sugiyama
- Department of Cardiology, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | | | - Peter Barlis
- Department of Medicine, St Vincent's Hospital, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Eric K W Poon
- Department of Medicine, St Vincent's Hospital, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Makoto Araki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Akihiro Nakajima
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yoshiyasu Minami
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Masamichi Takano
- Cardiovascular Center, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Chiba, Japan
| | - Osamu Kurihara
- Cardiovascular Center, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Chiba, Japan
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Tsunekazu Kakuta
- Department of Cardiology, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Cardiology Division, Kyung Hee University Hospital, Seoul, South Korea.
| |
Collapse
|
33
|
Dynamic Crosstalk between Vascular Smooth Muscle Cells and the Aged Extracellular Matrix. Int J Mol Sci 2021; 22:ijms221810175. [PMID: 34576337 PMCID: PMC8468233 DOI: 10.3390/ijms221810175] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Vascular aging is accompanied by the fragmentation of elastic fibers and collagen deposition, leading to reduced distensibility and increased vascular stiffness. A rigid artery facilitates elastin to degradation by MMPs, exposing vascular cells to greater mechanical stress and triggering signaling mechanisms that only exacerbate aging, creating a self-sustaining inflammatory environment that also promotes vascular calcification. In this review, we highlight the role of crosstalk between smooth muscle cells and the vascular extracellular matrix (ECM) and how aging promotes smooth muscle cell phenotypes that ultimately lead to mechanical impairment of aging arteries. Understanding the underlying mechanisms and the role of associated changes in ECM during aging may contribute to new approaches to prevent or delay arterial aging and the onset of cardiovascular diseases.
Collapse
|
34
|
Sun C, He B, Sun M, Lv X, Wang F, Chen J, Zhang J, Ye Z, Wen J, Liu P. Yes-Associated Protein in Atherosclerosis and Related Complications: A Potential Therapeutic Target That Requires Further Exploration. Front Cardiovasc Med 2021; 8:704208. [PMID: 34513949 PMCID: PMC8430249 DOI: 10.3389/fcvm.2021.704208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis and its complications diseases remain leading causes of cardiovascular morbidity and mortality, bringing a massive burden on public health worldwide. Atherosclerosis is recognized as chronic inflammation, and involves several highly correlated processes, including lipid metabolism dysfunction, endothelial cell dysfunction, inflammation, oxidative stress, vascular smooth muscle cell activation, platelet activation, thrombosis, altered matrix metabolism, and vascular remodeling. Within the past few decades, accumulating evidence has shown that the Yes-associated protein (YAP), the major effector of the Hippo pathway, can play a crucial role in pathogenesis and development of atherosclerosis. Activation of YAP-related pathways, which are induced by alerting flow pattern and matrix stiffness among others, can regulate processes including vascular endothelial cell dysfunction, monocyte infiltration, and smooth muscle cell migration, which contribute to atherosclerotic lesion formation. Further, YAP potentially modulates atherosclerotic complications such as vascular calcification and intraplaque hemorrhage, which require further investigation. Here, we summarized the relevant literature to outline current findings detailing the relationship between of YAP and atherosclerosis and highlight areas for future research.
Collapse
Affiliation(s)
- Congrui Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Bin He
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Mingsheng Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianbin Zhang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianyan Wen
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
35
|
Stula I, Kojundzic SL, Guic MM, Novak K. Carotid artery stenosis in correlation with neck and carotid artery anatomy. Vascular 2021; 30:524-531. [PMID: 34053369 DOI: 10.1177/17085381211018603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The purpose of this study was to examine the relationship between neck anatomy, especially its largest muscle - sternocleidomastoid and carotid space, with carotid artery anatomy and stenosis. METHODS We analysed 102 computed tomography carotid angiograms. The study included the measurement of the neck and sternocleidomastoid length, diameter and volume and the size of the carotid space. Analysis of carotid artery geometry, the length, angle and height of carotid artery bifurcation and the direction of the internal carotid artery origin was also included. RESULTS We found a positive correlation only between the neck and carotid length. There was no correlation between other neck characteristics and a carotid anatomy or internal carotid artery stenosis. Direction of internal carotid artery origin was significantly different (p < 0.01) between the left and right sides. CONCLUSIONS We have not found a correlation between the size of sternocleidomastoid and carotid space and carotid stenosis as a hypothetical factor for atherosclerosis. Also, the degree of carotid artery stenosis did not correlate with other neck and carotid measurements. Neck and carotid anatomy correlated only in their lengths. The left internal carotid artery showed mostly posterolateral origin, and right internal carotid artery had no predominate direction.
Collapse
Affiliation(s)
- Ivana Stula
- Department of Diagnostic and Interventional Radiology, University Hospital of Split, Split, Croatia.,School of Medicine, University of Split, Split, Croatia.,Department of Health Studies, University of Split, Split, Croatia
| | - Sanja L Kojundzic
- Department of Diagnostic and Interventional Radiology, University Hospital of Split, Split, Croatia.,School of Medicine, University of Split, Split, Croatia.,Department of Health Studies, University of Split, Split, Croatia
| | - Maja M Guic
- Department of Diagnostic and Interventional Radiology, University Hospital of Split, Split, Croatia.,Department of Health Studies, University of Split, Split, Croatia
| | | |
Collapse
|
36
|
Kheradvar A, Vannan MA, Dasi LP, Pedrizzetti G. The effect of aortic root anatomy and vortex flow induced shear stress on the aortic valve leaflets. Eur Heart J Cardiovasc Imaging 2021; 22:995-997. [PMID: 33758910 DOI: 10.1093/ehjci/jeab031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Arash Kheradvar
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, Department of Biomedical Enginnering, University of California, Irvine, CA, USA
| | - Mani A Vannan
- Marcus Heart Valve Center, Piedmont Heart Institute, Atlanta, GA, USA
| | - Lakshmi P Dasi
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gianni Pedrizzetti
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| |
Collapse
|
37
|
Tanaka K, Joshi D, Timalsina S, Schwartz MA. Early events in endothelial flow sensing. Cytoskeleton (Hoboken) 2021; 78:217-231. [PMID: 33543538 DOI: 10.1002/cm.21652] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Responses of vascular and lymphatic endothelial cells (ECs) to fluid shear stress (FSS) from blood or lymphatic fluid flow govern the development, physiology, and diseases of these structures. Extensive research has characterized the signaling, gene expression and cytoskeletal pathways that mediate effects on EC phenotype and vascular morphogenesis. But the primary mechanisms by which ECs transduce the weak forces from flow into biochemical signals are less well understood. This review covers recent advances in our understanding of the immediate mechanisms of FSS mechanotransduction, integrating results from different disciplines, addressing their roles in development, physiology and disease, and suggesting important questions for future work.
Collapse
Affiliation(s)
- Keiichiro Tanaka
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Divyesh Joshi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Sushma Timalsina
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA.,Department of Cell Biology, Yale University, New Haven, Connecticut, USA.,Department of Biomedical engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
38
|
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
|
39
|
Yamada S, Ito H, Ishikawa M, Yamamoto K, Yamaguchi M, Oshima M, Nozaki K. Quantification of Oscillatory Shear Stress from Reciprocating CSF Motion on 4D Flow Imaging. AJNR Am J Neuroradiol 2021; 42:479-486. [PMID: 33478942 DOI: 10.3174/ajnr.a6941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/05/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND PURPOSE Oscillatory shear stress could not be directly measured in consideration of direction, although cerebrospinal fluid has repetitive movements synchronized with heartbeat. Our aim was to evaluate the important of oscillatory shear stress in the cerebral aqueduct and foramen magnum in idiopathic normal pressure hydrocephalus by comparing it with wall shear stress and the oscillatory shear index in patients with idiopathic normal pressure hydrocephalus. MATERIALS AND METHODS By means of the 4D flow application, oscillatory shear stress, wall shear stress, and the oscillatory shear index were measured in 41 patients with idiopathic normal pressure hydrocephalus, 23 with co-occurrence of idiopathic normal pressure hydrocephalus and Alzheimer-type dementia, and 9 age-matched controls. These shear stress parameters at the cerebral aqueduct were compared with apertures and stroke volumes at the foramen of Magendie and cerebral aqueduct. RESULTS Two wall shear stress magnitude peaks during a heartbeat were changed to periodic oscillation by converting oscillatory shear stress. The mean oscillatory shear stress amplitude and time-averaged wall shear stress values at the dorsal and ventral regions of the cerebral aqueduct in the idiopathic normal pressure hydrocephalus groups were significantly higher than those in controls. Furthermore, those at the ventral region of the cerebral aqueduct in the idiopathic normal pressure hydrocephalus group were also significantly higher than those in the co-occurrence of idiopathic normal pressure hydrocephalus with Alzheimer-type dementia group. The oscillatory shear stress amplitude at the dorsal region of the cerebral aqueduct was significantly associated with foramen of Magendie diameters, whereas it was strongly associated with the stroke volume at the upper end of the cerebral aqueduct rather than that at the foramen of Magendie. CONCLUSIONS Oscillatory shear stress, which reflects wall shear stress vector changes better than the conventional wall shear stress magnitude and the oscillatory shear index, can be directly measured on 4D flow MR imaging. Oscillatory shear stress at the cerebral aqueduct was considerably higher in patients with idiopathic normal pressure hydrocephalus.
Collapse
Affiliation(s)
- S Yamada
- From the Department of Neurosurgery (S.Y., K.N.), Shiga University of Medical Science, Shiga, Japan .,Department of Neurosurgery and Normal Pressure Hydrocephalus Center (S.Y., K.Y., M.Y.), Rakuwakai Otowa Hospital, Kyoto, Japan.,Interfaculty Initiative in Information Studies/Institute of Industrial Science (S.Y., M.O.), The University of Tokyo, Tokyo, Japan
| | - H Ito
- Medical System Research and Development Center (H.I.), Fujifilm Corporation, Tokyo, Japan
| | | | - K Yamamoto
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center (S.Y., K.Y., M.Y.), Rakuwakai Otowa Hospital, Kyoto, Japan
| | - M Yamaguchi
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center (S.Y., K.Y., M.Y.), Rakuwakai Otowa Hospital, Kyoto, Japan
| | - M Oshima
- Interfaculty Initiative in Information Studies/Institute of Industrial Science (S.Y., M.O.), The University of Tokyo, Tokyo, Japan
| | - K Nozaki
- From the Department of Neurosurgery (S.Y., K.N.), Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
40
|
Teng Z, Wang S, Tokgoz A, Taviani V, Bird J, Sadat U, Huang Y, Patterson AJ, Figg N, Graves MJ, Gillard JH. Study on the association of wall shear stress and vessel structural stress with atherosclerosis: An experimental animal study. Atherosclerosis 2021; 320:38-46. [PMID: 33524908 DOI: 10.1016/j.atherosclerosis.2021.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Artery is subject to wall shear stress (WSS) and vessel structural stress (VSS) simultaneously. This study is designed to explore the role of VSS in development of atherosclerosis. METHODS Silastic collars were deployed on the carotid to create two constrictions on 13 rabbits for a distinct mechanical environment at the constriction. MRI was performed to visualize arteries' configuration. Animals with high fat (n = 9; Model-group) and normal diet (n = 4; Control-group) were sacrificed after 16 weeks. 3D fluid-structure interaction analysis was performed to quantify WSS and VSS simultaneously. RESULTS Twenty plaques were found in Model-group and 3 in Control-group. In Model-group, 8 plaques located proximally to the first constriction (Region-1, close to the heart) and 7 distally to the second (Region-2, close to the head) and 5 plaques were found on the contralateral side of 3 rabbits. Plaques at Region-1 tended to be bigger than those at Region-2 and the macrophage density at these locations was comparable. Minimum time-averaged WSS (TAWSS) in Region-1 was significantly higher than that in Region-2, and both maximum oscillatory shear index (OSI) and particle relative residence time (RRT) were significantly lower. Peak and mean VSS in Region-1 were significantly higher than those in Region-2. Correlation analyses indicated that low TAWSS, high OSI and RRT were only associated with plaque in Region-2, while lesions in Region-1 were only associated with high VSS. Moreover, only VSS was associated with wall thickness of plaque-free regions in both regions. CONCLUSIONS VSS might contribute to the initialization and development of atherosclerosis solely or in combination with WSS.
Collapse
Affiliation(s)
- Zhongzhao Teng
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Department of Engineering, University of Cambridge, Cambridge, United Kingdom.
| | - Shuo Wang
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Aziz Tokgoz
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Valentina Taviani
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Bird
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Umar Sadat
- Cambridge Vascular Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Yuan Huang
- EPSRC Centre for Mathematical and Statistical Analysis of Multimodal Clinical Imaging, University of Cambridge, Cambridge, United Kingdom
| | - Andrew J Patterson
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Nichola Figg
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan H Gillard
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
41
|
Yufa A, Mikael A, Lara G, Nurick H, Andacheh I. Accessory renal arteries involved in atherosclerotic occlusive disease at the aortic bifurcation. JOURNAL OF VASCULAR SURGERY CASES INNOVATIONS AND TECHNIQUES 2020; 6:425-429. [PMID: 33367190 PMCID: PMC7748983 DOI: 10.1016/j.jvscit.2020.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/02/2020] [Indexed: 11/28/2022]
Abstract
Accessory renal arteries (ARAs) are embryonic remnants found in more than one-third of patients and occurring bilaterally in 10% of the population. Very few reports have documented such vessels arising near or at the level of the aortic bifurcation. Furthermore, the presence of ARAs has yet to be described in the context of atherosclerotic disease. Here, we present a unique case of large bilateral ARAs originating above the aortic bifurcation concurrent with symptomatic aortoiliac atherosclerotic disease. We highlight the embryologic and clinical significance of these vessels as well as discuss their potential role in accelerating atherosclerotic disease processes.
Collapse
Affiliation(s)
- Ann Yufa
- University of California, Riverside School of Medicine, Riverside, Calif.,Riverside Community Hospital, Riverside, Calif
| | | | - Gustavo Lara
- Riverside Community Hospital, Riverside, Calif.,Harvey Nurick, MD, Inc, Riverside, Calif
| | - Harvey Nurick
- Riverside Community Hospital, Riverside, Calif.,Harvey Nurick, MD, Inc, Riverside, Calif
| | - Iden Andacheh
- University of California, Riverside School of Medicine, Riverside, Calif.,Riverside Community Hospital, Riverside, Calif.,Harvey Nurick, MD, Inc, Riverside, Calif
| |
Collapse
|
42
|
McCallinhart PE, Scandling BW, Trask AJ. Coronary remodeling and biomechanics: Are we going with the flow in 2020? Am J Physiol Heart Circ Physiol 2020; 320:H584-H592. [PMID: 33185115 DOI: 10.1152/ajpheart.00634.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Under normal conditions, coronary blood flow (CBF) provides critical blood supply to the myocardium so that it can appropriately meet the metabolic demands of the body. Dogmatically, there exist several known regulators and modulators of CBF that include local metabolites and neurohormonal factors that can influence the function of the coronary circulation. In disease states such as diabetes and myocardial ischemia, these regulators are impaired or shifted such that CBF is reduced. Although functional considerations have been and continued to be well studied, more recent evidence builds upon established studies that collectively suggest that the relative roles of coronary structure, biomechanics, and the influence of cardiac biomechanics via extravascular compression may also play a significant role in dictating CBF. In this mini review, we discuss these regulators of CBF under normal and pathophysiological conditions and their potential influence on the control of CBF.
Collapse
Affiliation(s)
- Patricia E McCallinhart
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
| | - Benjamin W Scandling
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio.,Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Aaron J Trask
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
43
|
Grégory Franck. Role of mechanical stress and neutrophils in the pathogenesis of plaque erosion. Atherosclerosis 2020; 318:60-69. [PMID: 33190807 DOI: 10.1016/j.atherosclerosis.2020.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/05/2020] [Accepted: 11/03/2020] [Indexed: 02/05/2023]
Abstract
Mechanical stress is a well-recognized driver of plaque rupture. Likewise, investigating the role of mechanical forces in plaque erosion has recently begun to provide some important insights, yet the knowledge is by far less advanced. The most significant example is that of shear stress, which has early been proposed as a possible driver for focal endothelial death and denudation. Recent findings using optical coherence tomography, computational sciences and mechanical models show that plaque erosion occurs most likely around atheromatous plaque throats with specific stress pattern. In parallel, we have recently shown that neutrophil-dependent inflammation promotes plaque erosion, possibly through a noxious action on ECs. Most importantly, spontaneous thrombosis - associated or not with EC denudation - can be impacted by hemodynamics, and it is now established that neutrophils promote thrombosis and platelet activation, highlighting a potential relationship between, mechanical stress, inflammation, and EC loss in the setting of coronary plaque erosion. Here, we review our current knowledge regarding the implication of both mechanical stress and neutrophils, and we discuss their implication in the promotion of plaque erosion via EC loss and thrombosis.
Collapse
Affiliation(s)
- Grégory Franck
- Inserm LVTS U1148. CHU Bichat, 46 Rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
44
|
Shedding 3D light on endothelial barrier function in arteries. Atherosclerosis 2020; 310:91-92. [DOI: 10.1016/j.atherosclerosis.2020.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/31/2020] [Indexed: 11/19/2022]
|
45
|
Karthika CL, Ahalya S, Radhakrishnan N, Kartha CC, Sumi S. Hemodynamics mediated epigenetic regulators in the pathogenesis of vascular diseases. Mol Cell Biochem 2020; 476:125-143. [PMID: 32844345 DOI: 10.1007/s11010-020-03890-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022]
Abstract
Endothelium of blood vessels is continuously exposed to various hemodynamic forces. Flow-mediated epigenetic plasticity regulates vascular endothelial function. Recent studies have highlighted the significant role of mechanosensing-related epigenetics in localized endothelial dysfunction and the regional susceptibility for lesions in vascular diseases. In this article, we review the epigenetic mechanisms such as DNA de/methylation, histone modifications, as well as non-coding RNAs in promoting endothelial dysfunction in major arterial and venous diseases, consequent to hemodynamic alterations. We also discuss the current challenges and future prospects for the use of mechanoepigenetic mediators as biomarkers of early stages of vascular diseases and dysregulated mechanosensing-related epigenetic regulators as therapeutic targets in various vascular diseases.
Collapse
Affiliation(s)
- C L Karthika
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - S Ahalya
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - N Radhakrishnan
- St.Thomas Institute of Research on Venous Diseases, Changanassery, Kerala, India
| | - C C Kartha
- Society for Continuing Medical Education & Research (SOCOMER), Kerala Institute of Medical Sciences, Thiruvananthapuram, Kerala, India
| | - S Sumi
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
46
|
Cameron JN, Mehta OH, Michail M, Chan J, Nicholls SJ, Bennett MR, Brown AJ. Exploring the relationship between biomechanical stresses and coronary atherosclerosis. Atherosclerosis 2020; 302:43-51. [PMID: 32438198 DOI: 10.1016/j.atherosclerosis.2020.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/05/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
The pathophysiology of coronary atherosclerosis is multifaceted. Plaque initiation and progression are governed by a complex interplay between genetic and environmental factors acting through processes such as lipid accumulation, altered haemodynamics and inflammation. There is increasing recognition that biomechanical stresses play an important role in atherogenesis, and integration of these metrics with clinical imaging has potential to significantly improve cardiovascular risk prediction. In this review, we present the calculation of coronary biomechanical stresses from first principles and computational methods, including endothelial shear stress (ESS), plaque structural stress (PSS) and axial plaque stress (APS). We discuss the current experimental and human data linking these stresses to the natural history of coronary artery disease and explore the future potential for refining treatment options and predicting future ischaemic events.
Collapse
Affiliation(s)
- James N Cameron
- Monash Cardiovascular Research Centre and MonashHeart, Monash Health, Melbourne, Australia
| | - Ojas H Mehta
- Monash Cardiovascular Research Centre and MonashHeart, Monash Health, Melbourne, Australia
| | - Michael Michail
- Monash Cardiovascular Research Centre and MonashHeart, Monash Health, Melbourne, Australia; Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Jasmine Chan
- Monash Cardiovascular Research Centre and MonashHeart, Monash Health, Melbourne, Australia
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre and MonashHeart, Monash Health, Melbourne, Australia
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Adam J Brown
- Monash Cardiovascular Research Centre and MonashHeart, Monash Health, Melbourne, Australia.
| |
Collapse
|
47
|
Zhang X, Ramírez CM, Aryal B, Madrigal-Matute J, Liu X, Diaz A, Torrecilla-Parra M, Suárez Y, Cuervo AM, Sessa WC, Fernández-Hernando C. Cav-1 (Caveolin-1) Deficiency Increases Autophagy in the Endothelium and Attenuates Vascular Inflammation and Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:1510-1522. [PMID: 32349535 PMCID: PMC7253189 DOI: 10.1161/atvbaha.120.314291] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Endothelial Cav-1 (caveolin-1) expression plays a relevant role during atherogenesis by controlling NO production, vascular inflammation, LDL (low-density lipoprotein) transcytosis, and extracellular matrix remodeling. Additional studies have identified cholesterol-rich membrane domains as important regulators of autophagy by recruiting ATGs (autophagy-related proteins) to the plasma membrane. Here, we investigate how the expression of Cav-1 in the aortic endothelium influences autophagy and whether enhanced autophagy contributes to the atheroprotective phenotype observed in Cav-1–deficient mice.
Collapse
Affiliation(s)
- Xinbo Zhang
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Cristina M Ramírez
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,IMDEA Research Institute of Food and Health Sciences, Madrid, Spain (C.M.R., M.T.-P.)
| | - Binod Aryal
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Julio Madrigal-Matute
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (J.M.-M., A.D., A.M.C.)
| | - Xinran Liu
- Department of Cell Biology (X.L.), Yale University School of Medicine, New Haven, CT
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (J.M.-M., A.D., A.M.C.)
| | | | - Yajaira Suárez
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Ana M Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (J.M.-M., A.D., A.M.C.)
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT
| | - Carlos Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
48
|
Piskin S, Patnaik SS, Han D, Bordones AD, Murali S, Finol EA. A canonical correlation analysis of the relationship between clinical attributes and patient-specific hemodynamic indices in adult pulmonary hypertension. Med Eng Phys 2020; 77:1-9. [PMID: 32007361 DOI: 10.1016/j.medengphy.2020.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/19/2019] [Accepted: 01/06/2020] [Indexed: 11/19/2022]
Abstract
Pulmonary hypertension (PH) is a progressive disease affecting approximately 10-52 cases per million, with a higher incidence in women, and with a high mortality associated with right ventricle (RV) failure. In this work, we explore the relationship between hemodynamic indices, calculated from in silico models of the pulmonary circulation, and clinical attributes of RV workload and pathological traits. Thirty-four patient-specific pulmonary arterial tree geometries were reconstructed from computed tomography angiography images and used for volume meshing for subsequent computational fluid dynamics (CFD) simulations. Data obtained from the CFD simulations were post-processed resulting in hemodynamic indices representative of the blood flow dynamics. A retrospective review of medical records was performed to collect the clinical variables measured or calculated from standard hospital examinations. Statistical analyses and canonical correlation analysis (CCA) were performed for the clinical variables and hemodynamic indices. Systolic pulmonary artery pressure (sPAP), diastolic pulmonary artery pressure (dPAP), cardiac output (CO), and stroke volume (SV) were moderately correlated with spatially averaged wall shear stress (0.60 ≤ R2 ≤ 0.66; p < 0.05). Similarly, the CCA revealed a linear and strong relationship (ρ = 0.87; p << 0.001) between 5 clinical variables and 2 hemodynamic indices. To this end, in silico models of PH blood flow dynamics have a high potential for predicting the relevant clinical attributes of PH if analyzed in a group-wise manner using CCA.
Collapse
Affiliation(s)
- Senol Piskin
- Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA; Department of Mechanical Engineering, Istinye University, Zeytinburnu, Istanbul 34010, Turkey
| | - Sourav S Patnaik
- Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA.
| | - David Han
- Department of Management Science and Statistics, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA.
| | - Alifer D Bordones
- Department of Biomedical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA.
| | - Srinivas Murali
- Department of Radiology and Department of Cardiology, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA.
| | - Ender A Finol
- Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA.
| |
Collapse
|
49
|
Tsompou PI, Siogkas PK, Sakellarios AI, Andrikos IO, Kigka VI, Lemos PA, Michalis LK, Fotiadis DI. A comparison of three multimodality coronary 3D reconstruction methods. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:5812-5815. [PMID: 31947173 DOI: 10.1109/embc.2019.8857582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The assessment of the severity of arterial stenoses is of utmost importance in clinical practice. Several image modalities invasive and non-invasive are nowadays available and can be utilized for the 3-dimensional (3D) reconstruction of the arterial geometry. Following our previous study, the present study was conducted to further strengthen the evaluation of three reconstruction methodologies, namely: (i) the Quantitative Coronary Analysis (QCA), (ii) the Virtual Histology Intravascular Ultrasound VH-IVUS-Angiography hybrid method and (iii) the Coronary Computed Tomography Angiography (CCTA). Data from 13 patients were employed to perform a quantitative analysis using specific metrics, such as, the Mean Wall Shear Stress (mWSS), the Minimum Lumen diameter (MLD), the Reference Vessel Diameter (RVD), the Degree of stenosis (DS%), and the Lesion length (LL). A high correlation was observed for the mWSS metric between the three reconstruction methods, especially between the QCA and CCTA (r=0.974, P<; 0.001).
Collapse
|
50
|
|