1
|
Bai H, Varsanik MA, Thaxton C, Ohashi Y, Gonzalez L, Zhang W, Aoyagi Y, Kano M, Yatsula B, Li Z, Pocivavsek L, Dardik A. Disturbed flow in the juxta-anastomotic area of an arteriovenous fistula correlates with endothelial loss, acute thrombus formation, and neointimal hyperplasia. Am J Physiol Heart Circ Physiol 2024; 326:H1446-H1461. [PMID: 38578237 PMCID: PMC11380968 DOI: 10.1152/ajpheart.00054.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Clinical failure of arteriovenous neointimal hyperplasia (NIH) fistulae (AVF) is frequently due to juxta-anastomotic NIH (JANIH). Although the mouse AVF model recapitulates human AVF maturation, previous studies focused on the outflow vein distal to the anastomosis. We hypothesized that the juxta-anastomotic area (JAA) has increased NIH compared with the outflow vein. AVF was created in C57BL/6 mice without or with chronic kidney disease (CKD). Temporal and spatial changes of the JAA were examined using histology and immunofluorescence. Computational techniques were used to model the AVF. RNA-seq and bioinformatic analyses were performed to compare the JAA with the outflow vein. The jugular vein to carotid artery AVF model was created in Wistar rats. The neointima in the JAA shows increased volume compared with the outflow vein. Computational modeling shows an increased volume of disturbed flow at the JAA compared with the outflow vein. Endothelial cells are immediately lost from the wall contralateral to the fistula exit, followed by thrombus formation and JANIH. Gene Ontology (GO) enrichment analysis of the 1,862 differentially expressed genes (DEG) between the JANIH and the outflow vein identified 525 overexpressed genes. The rat jugular vein to carotid artery AVF showed changes similar to the mouse AVF. Disturbed flow through the JAA correlates with rapid endothelial cell loss, thrombus formation, and JANIH; late endothelialization of the JAA channel correlates with late AVF patency. Early thrombus formation in the JAA may influence the later development of JANIH.NEW & NOTEWORTHY Disturbed flow and focal endothelial cell loss in the juxta-anastomotic area of the mouse AVF colocalizes with acute thrombus formation followed by late neointimal hyperplasia. Differential flow patterns between the juxta-anastomotic area and the outflow vein correlate with differential expression of genes regulating coagulation, proliferation, collagen metabolism, and the immune response. The rat jugular vein to carotid artery AVF model shows changes similar to the mouse AVF model.
Collapse
MESH Headings
- Animals
- Neointima
- Hyperplasia
- Arteriovenous Shunt, Surgical
- Thrombosis/physiopathology
- Thrombosis/pathology
- Thrombosis/genetics
- Thrombosis/etiology
- Thrombosis/metabolism
- Mice, Inbred C57BL
- Rats, Wistar
- Male
- Jugular Veins/metabolism
- Jugular Veins/pathology
- Jugular Veins/physiopathology
- Disease Models, Animal
- Carotid Arteries/pathology
- Carotid Arteries/physiopathology
- Carotid Arteries/metabolism
- Carotid Arteries/surgery
- Mice
- Rats
- Regional Blood Flow
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/pathology
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/physiopathology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
Collapse
Affiliation(s)
- Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - M Alyssa Varsanik
- Section of Vascular Surgery, Department of Surgery, University of Chicago Medicine, Chicago, Illinois, United States
| | - Carly Thaxton
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yukihiko Aoyagi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Masaki Kano
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Zhuo Li
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Luka Pocivavsek
- Section of Vascular Surgery, Department of Surgery, University of Chicago Medicine, Chicago, Illinois, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Cellular and Molecular Physiology, Yale University; New Haven, Connecticut, United States
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut, United States
| |
Collapse
|
2
|
Kalinin RY, Suсhkov IA, Klimentova EA, Shanaev IN. The effect of the geometry of the proximal anastomosis, markers of apoptosis and cell proliferation on the long-term patency following reconstructive interventions on the femoropopliteal arterial segment. KAZAN MEDICAL JOURNAL 2021; 102:855-861. [DOI: 10.17816/kmj2021-855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Aim. To study changes in the topography of the orifice of the deep femoral artery (DFA), markers of proliferation, and apoptosis in patients after open interventions on the femoropopliteal arterial segment.
Methods. The study included 35 patients with atherosclerotic peripheral arterial disease (PAD), femoral-popliteal occlusion, stage IIBIII of the disease according to the classification of A.V. PokrovskyFontaine, who underwent open surgery. The average age of the patients was 694.6 years. These patients included 26 men. Patients were divided into two groups: group A included 18 patients who underwent femoral-popliteal prosthetics (distal End-To-End bypass anastomoses), group B included 17 patients with femoral-popliteal bypass surgery (distal End-To-Side bypass anastomoses). The groups were comparable in terms of age and disease severity (p 0.05). Determination of serum platelet-derived growth factor BB (PDGF BB) and soluble form Fas (sFas) levels was carried out immediately before the intervention, on the 1st, 7th days, and 1 month after the operation. Duplex scanning (DS) was performed on day 7, after 1 and 18 months. Statistica 10.0 software was used for statistical data processing. The significance of differences between unrelated samples was assessed using the Student's t-test. The correlations between variables were analyzed by using Pearson's method.
Results. On 1st day, there was a decrease in soluble Fas in patients of group A compared with group B (0.41 ng/ml vs 0.78 ng/ml, p=0.01). On the 7th day, the levels of serum platelet-derived growth factor BB were increased in patients of group A compared with group B (35.2 ng/ml vs 23.2 ng/ml, p=0.00001). After 1 month, the level of serum platelet-derived growth factor BB in patients of group A remained elevated compared with those in patients of group B (22.8 ng/ml vs 14.4 ng/ml, p=0.0003).
Conclusion. Femoropopliteal prosthetics leads to a change in branching angle of the deep femoral artery up to 7080%, accompanied by changing dynamics of apoptotic markers and cell proliferation, leading to an increase in the thickness of neointimal hyperplasia and the progression of atherosclerosis.
Collapse
|
3
|
Yang CY, Chang PY, Wu BS, Tarng DC, Lee OKS. Mechanical and chemical cues synergistically promote human venous smooth muscle cell osteogenesis through integrin β1-ERK1/2 signaling: A cell model of hemodialysis fistula calcification. FASEB J 2021; 35:e22042. [PMID: 34758125 DOI: 10.1096/fj.202101064rr] [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: 06/24/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 11/11/2022]
Abstract
Arteriovenous fistula (AVF) is the vascular access of choice for renal replacement therapy. However, AVF is susceptible to calcification with a high prevalence of 40%-65% in chronic hemodialysis patients. Repeated needle puncture for hemodialysis cannulation results in intimal denudation of AVF. We hypothesized that exposure to blood shear stress in the medial layer promotes venous smooth muscle cell (SMC) osteogenesis. While previous studies of shear stress focused on arterial-type SMCs, SMCs isolated from the vein had not been investigated. This study established a venous cell model of AVF using the fluid shear device, combined with a high phosphate medium to mimic the uremic milieu. Osteogenic gene expression of venous SMCs upon mechanical and chemical cues was analyzed in addition to the activated cell signaling pathways. Our findings indicated that upon shear stress and high phosphate environment, mechanical stimulation (shear stress) had an additive effect in up-regulation of an early osteogenic marker, Runx2. We further identified that the integrin β1-ERK1/2 signaling pathway was responsible for the molecular basis of venous SMC osteogenesis upon shear stress exposure. Mitochondrial biogenesis also took part in the early stage of this venopathy pathogenesis, evident by the up-regulated mitochondrial transcription factor A and mitochondrial DNA polymerase γ in venous SMCs. In conclusion, synergistic effects of fluid shear stress and high phosphate induce venous SMC osteogenesis via the ERK1/2 pathway through activating the mechanosensing integrin β1 signaling. The present study identified a promising druggable target for reducing AVF calcification, which deserves further in vivo investigations.
Collapse
Affiliation(s)
- Chih-Yu Yang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), Ministry of Education, Taipei, Taiwan.,Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pu-Yuan Chang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Bo-Sheng Wu
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), Ministry of Education, Taipei, Taiwan
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Jensen LF, Bentzon JF, Albarrán-Juárez J. The Phenotypic Responses of Vascular Smooth Muscle Cells Exposed to Mechanical Cues. Cells 2021; 10:2209. [PMID: 34571858 PMCID: PMC8469800 DOI: 10.3390/cells10092209] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
During the development of atherosclerosis and other vascular diseases, vascular smooth muscle cells (SMCs) located in the intima and media of blood vessels shift from a contractile state towards other phenotypes that differ substantially from differentiated SMCs. In addition, these cells acquire new functions, such as the production of alternative extracellular matrix (ECM) proteins and signal molecules. A similar shift in cell phenotype is observed when SMCs are removed from their native environment and placed in a culture, presumably due to the absence of the physiological signals that maintain and regulate the SMC phenotype in the vasculature. The far majority of studies describing SMC functions have been performed under standard culture conditions in which cells adhere to a rigid and static plastic plate. While these studies have contributed to discovering key molecular pathways regulating SMCs, they have a significant limitation: the ECM microenvironment and the mechanical forces transmitted through the matrix to SMCs are generally not considered. Here, we review and discuss the recent literature on how the mechanical forces and derived biochemical signals have been shown to modulate the vascular SMC phenotype and provide new perspectives about their importance.
Collapse
Affiliation(s)
- Lise Filt Jensen
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark; (L.F.J.); (J.F.B.)
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark; (L.F.J.); (J.F.B.)
- Experimental Pathology of Atherosclerosis Laboratory, Spanish National Center for Cardiovascular Research (CNIC), 28029 Madrid, Spain
- Steno Diabetes Center Aarhus, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Julian Albarrán-Juárez
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark; (L.F.J.); (J.F.B.)
| |
Collapse
|
5
|
Hartman EMJ, De Nisco G, Kok AM, Hoogendoorn A, Coenen A, Mastik F, Korteland SA, Nieman K, Gijsen FJH, van der Steen AFW, Daemen J, Wentzel JJ. Lipid-rich Plaques Detected by Near-infrared Spectroscopy Are More Frequently Exposed to High Shear Stress. J Cardiovasc Transl Res 2020; 14:416-425. [PMID: 33034862 PMCID: PMC8219563 DOI: 10.1007/s12265-020-10072-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 09/14/2020] [Indexed: 01/06/2023]
Abstract
High wall shear stress (WSS) and near-infrared spectroscopy (NIRS) detected lipid-rich plaque (LRP) are both known to be associated with plaque destabilization and future adverse cardiovascular events. However, knowledge of spatial co-localization of LRP and high WSS is lacking. This study investigated the co-localization of LRP based on NIRS and high WSS. Fifty-three patients presenting acute coronary syndrome underwent NIRS-intravascular-ultrasound (NIRS-IVUS) imaging of a non-culprit coronary artery. WSS was obtained using WSS profiling in 3D-reconstructions of the coronary arteries based on fusion of IVUS-segmented lumen and CT-derived 3D-centerline. Thirty-eight vessels were available for final analysis and divided into 0.5 mm/45° sectors. LRP sectors, as identified by NIRS, were more often colocalized with high WSS than sectors without LRP. Moreover, there was a dose-dependent relationship between lipid content and high WSS exposure. This study is a first step in understanding the evolution of LRPs to vulnerable plaques. Graphical Abstract ![]()
Collapse
Affiliation(s)
| | - Giuseppe De Nisco
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Annette M Kok
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Ayla Hoogendoorn
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Adriaan Coenen
- Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| | - Frits Mastik
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Koen Nieman
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands.,Department of Radiology, Erasmus MC, Rotterdam, The Netherlands.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Frank J H Gijsen
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Joost Daemen
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
6
|
Cell signaling model for arterial mechanobiology. PLoS Comput Biol 2020; 16:e1008161. [PMID: 32834001 PMCID: PMC7470387 DOI: 10.1371/journal.pcbi.1008161] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/03/2020] [Accepted: 07/17/2020] [Indexed: 11/20/2022] Open
Abstract
Arterial growth and remodeling at the tissue level is driven by mechanobiological processes at cellular and sub-cellular levels. Although it is widely accepted that cells seek to promote tissue homeostasis in response to biochemical and biomechanical cues—such as increased wall stress in hypertension—the ways by which these cues translate into tissue maintenance, adaptation, or maladaptation are far from understood. In this paper, we present a logic-based computational model for cell signaling within the arterial wall, aiming to predict changes in extracellular matrix turnover and cell phenotype in response to pressure-induced wall stress, flow-induced wall shear stress, and exogenous sources of angiotensin II, with particular interest in mouse models of hypertension. We simulate a number of experiments from the literature at both the cell and tissue level, involving single or combined inputs, and achieve high qualitative agreement in most cases. Additionally, we demonstrate the utility of this modeling approach for simulating alterations (in this case knockdowns) of individual nodes within the signaling network. Continued modeling of cellular signaling will enable improved mechanistic understanding of arterial growth and remodeling in health and disease, and will be crucial when considering potential pharmacological interventions. Biological soft tissues are characterized by continuous production and removal of material, which endows them with a remarkable ability to adapt to changes in their biochemical and biomechanical environments. For arteries, mechanical stimuli result primarily from changes in blood pressure or flow, and biochemical changes are induced by multiple factors, including pharmacological intervention. In order to understand how arterial properties are maintained in health, or how they adapt or fail to adapt in disease, we must understand better how these diverse stimuli affect material turnover. Extracellular matrix is tightly regulated by mechano-sensing and mechano-regulation, and therefore cell signaling, thus we present a computational model of relevant signaling pathways within the vascular wall, with the aim of predicting changes in wall composition and function in response to three main inputs: pressure-induced wall stress, flow-induced wall shear stress, and exogenous angiotensin II. We obtain qualitative agreement with a range of experimental studies from the literature, and provide illustrative examples demonstrating how such models can be used to further our understanding of arterial remodeling.
Collapse
|
7
|
Chen J, Zhou Y, Liu S, Li C. Biomechanical signal communication in vascular smooth muscle cells. J Cell Commun Signal 2020; 14:357-376. [PMID: 32780323 DOI: 10.1007/s12079-020-00576-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Biomechanical stresses are closely associated with cardiovascular development and diseases. In vivo, vascular smooth muscle cells are constantly stimulated by biomechanical factors caused by increased blood pressure leading to the non-specific activation of cell transmembrane proteins. Thus, various intracellular signal molecules are simultaneously activated via signaling cascades, which are closely related to alterations in the differentiation, phenotype, inflammation, migration, pyroptosis, calcification, proliferation, and apoptosis of vascular smooth muscle cells. Meanwhile, mechanical stress-induced miRNAs and epigenetics modification on vascular smooth muscle cells play critical roles as well. Eventually, the overall pathophysiology of the cells is altered, resulting in the development of many major clinical diseases, including hypertension, atherosclerosis, grafted venous atherosclerosis, and aneurysm, among others. In this paper, important advances in mechanical signal communication in vascular smooth muscle cells are reviewed.
Collapse
Affiliation(s)
- Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Kalinin RE, Suchkov IA, Klimentova ЕA, Egorov AA, Povarov VO. Apoptosis in vascular pathology: present and future. I.P. PAVLOV RUSSIAN MEDICAL BIOLOGICAL HERALD 2020; 28:79-87. [DOI: 10.23888/pavlovj202028179-87] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Apoptosis is recognized as a programmed cell death controlled by genetic mechanisms and required for normal existence of an organism. Its main task is elimination of defective or mutant cells. The particles of dead cells are engulfed by macrophages with no development of inflammatory reaction. Apoptosis actively participates in embryogenesis, cellular homeostasis, elimination of tumor cells, and may be divided to three phases: signal, effector, and degradation. Its main components are cytoplasmic proteases caspases. Caspases exist in the cytoplasm in inactive condition in the form pf procaspases. Being activated, they break down to subunits. Proteins of Bcl-2 family are active participants of the mitochondrial pathway of apoptosis. They influence permeability of the outer membrane of mitochondria. Disorders in the mechanisms of apoptosis underlie many diseases including ischemic lesions, autoimmune disorders, malignant neoplasms. The ability to influence survival or death of cell is known to possess enormous therapeutic potential. At present, active research is under way to study signal pathways that control cell cycle and apoptosis. The article discusses the mechanisms participating in death of vascular endothelium and smooth muscle cells, potential role of apoptosis in atherosclerosis is also described.
Collapse
|
9
|
High shear stress on the coronary arterial wall is related to computed tomography-derived high-risk plaque: a three-dimensional computed tomography and color-coded tissue-characterizing intravascular ultrasonography study. Heart Vessels 2019; 34:1429-1439. [PMID: 30976923 DOI: 10.1007/s00380-019-01389-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/22/2019] [Indexed: 10/27/2022]
Abstract
Low wall shear stress (WSS) is associated with plaque formation. However, the relationship between WSS and coronary plaque vulnerability remains unclear. Therefore, this study aimed to clarify the in vivo relationship between luminal WSS derived from three-dimensional (3D) computed tomography (CT) and plaque vulnerability within the coronary artery. Forty-three consecutive patients with ischemic heart disease and coronary stenotic lesions were enrolled and underwent coronary angiography and color-coded intravascular ultrasonography (iMap™) followed by multi-slice coronary CT angiography. CT-derived high-risk plaque was defined by specific CT characteristics, including low CT intensity (< 30 HU) and positive remodeling. The Student's t test, Mann-Whitney U test, χ2 test, repeated measures analysis of variance, and logistic and multiple regression were used for statistical analyses. CT-derived high-risk plaque (n = 15) had higher values of maximum and average shear stress than CT-derived stable plaque (474 ± 453 vs. 158 ± 138 Pa, p = 0.018; 4.2 ± 3.1 vs. 1.6 ± 1.2 Pa, p = 0.007, respectively). Compared with patients with CT-derived stable plaque, those with CT-derived high-risk plaque had a higher prevalence of necrotic and lipidic characteristics (44 ± 13 vs. 31 ± 11%, p = 0.001) based on iMap™. Multivariate logistic regression analysis showed that the average WSS and necrotic plus lipidic content were independent determinants of CT-derived high-risk plaque (average WSS: odds ratio 2.996, p = 0.014; necrotic plus lipidic content: odds ratio 1.306, p = 0.036). Our findings suggested that CT-derived high-risk plaque may coexist with high shear stress on the plaque surface.
Collapse
|
10
|
Luo H, Zhou C, Chi J, Pan S, Lin H, Gao F, Ni T, Meng L, Zhang J, Jiang C, Ji Z, Lv H, Guo H. The Role of Tauroursodeoxycholic Acid on Dedifferentiation of Vascular Smooth Muscle Cells by Modulation of Endoplasmic Reticulum Stress and as an Oral Drug Inhibiting In-Stent Restenosis. Cardiovasc Drugs Ther 2019; 33:25-33. [DOI: 10.1007/s10557-018-6844-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
11
|
da Silva RA, Fernandes CJDC, Feltran GDS, Gomes AM, Andrade AF, Andia DC, Peppelenbosch MP, Zambuzzi WF. Laminar shear stress‐provoked cytoskeletal changes are mediated by epigenetic reprogramming of
TIMP1
in human primary smooth muscle cells. J Cell Physiol 2018; 234:6382-6396. [DOI: 10.1002/jcp.27374] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/17/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Rodrigo A. da Silva
- Department of Chemistry and Biochemistry Laboratory of Bioassays and Cellular Dynamics, São Paulo State University (UNESP), Institute of Biosciences, Campus Botucatu Botucatu Brazil
| | - Célio Jr da C. Fernandes
- Department of Chemistry and Biochemistry Laboratory of Bioassays and Cellular Dynamics, São Paulo State University (UNESP), Institute of Biosciences, Campus Botucatu Botucatu Brazil
| | - Geórgia da S. Feltran
- Department of Chemistry and Biochemistry Laboratory of Bioassays and Cellular Dynamics, São Paulo State University (UNESP), Institute of Biosciences, Campus Botucatu Botucatu Brazil
| | - Anderson M. Gomes
- Department of Chemistry and Biochemistry Laboratory of Bioassays and Cellular Dynamics, São Paulo State University (UNESP), Institute of Biosciences, Campus Botucatu Botucatu Brazil
| | - Amanda Fantini Andrade
- Department of Chemistry and Biochemistry Laboratory of Bioassays and Cellular Dynamics, São Paulo State University (UNESP), Institute of Biosciences, Campus Botucatu Botucatu Brazil
| | - Denise C. Andia
- Faculdade de Odontologia Área de Pesquisa em Epigenética, Universidade Paulista, UNIP São Paulo São Paulo Brazil
| | - Maikel P. Peppelenbosch
- Department of Gastroenterology & Hepatology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
| | - Willian F. Zambuzzi
- Department of Chemistry and Biochemistry Laboratory of Bioassays and Cellular Dynamics, São Paulo State University (UNESP), Institute of Biosciences, Campus Botucatu Botucatu Brazil
- Electron Microscopy Center, São Paulo State University (UNESP), Institute of Biosciences, campus Botucatu Botucatu Brazil
| |
Collapse
|
12
|
Pan X, Wang B, Yuan T, Zhang M, Kent KC, Guo LW. Analysis of Combined Transcriptomes Identifies Gene Modules that Differentially Respond to Pathogenic Stimulation of Vascular Smooth Muscle and Endothelial Cells. Sci Rep 2018; 8:395. [PMID: 29321689 PMCID: PMC5762668 DOI: 10.1038/s41598-017-18675-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/15/2017] [Indexed: 12/22/2022] Open
Abstract
Smooth muscle cells (SMCs) and endothelial cells (ECs) are vital cell types composing the vascular medial wall and the atheroprotective inner lining, respectively. Current treatments for cardiovascular disease inhibit SMC hyperplasia but compromise EC integrity, predisposing patients to thrombosis. Therapeutics targeting SMCs without collateral damage to ECs are highly desirable. However, differential (SMC versus EC) disease-associated regulations remain poorly defined. We conducted RNA-seq experiments to investigate SMC-versus-EC differential transcriptomic dynamics, following treatment of human primary SMCs and ECs with TNFα or IL-1β, both established inducers of SMC hyperplasia and EC dysfunction. As revealed by combined SMC/EC transcriptomes, after TNFα or IL-1β induction, 174 and 213 genes respectively showed greater up-regulation in SMCs than in ECs (SMC-enriched), while 117 and 138 genes showed greater up-regulation in ECs over SMCs (EC-enriched). Analysis of gene interaction networks identified central genes shared in the two SMC-enriched gene sets, and a distinct group of central genes common in the two EC-enriched gene sets. Significantly, four gene modules (subnetworks) were identified from these central genes, including SMC-enriched JUN and FYN modules and EC-enriched SMAD3 and XPO1 modules. These modules may inform potential intervention targets for selective blockage of SMC hyperplasia without endothelial damage.
Collapse
Affiliation(s)
- Xiaokang Pan
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,James Molecular Laboratory, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43240, USA
| | - Bowen Wang
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Tiezheng Yuan
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Mengxue Zhang
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Surgery and Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - K Craig Kent
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Lian-Wang Guo
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA. .,Department of Surgery and Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
13
|
Zhang Z, Chai W, Xiong R, Zhou L, Huang Y. Printing-induced cell injury evaluation during laser printing of 3T3 mouse fibroblasts. Biofabrication 2017. [DOI: 10.1088/1758-5090/aa6ed9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
14
|
Zhang Z, Chai W, Xiong R, Zhou L, Huang Y. Printing-induced cell injury evaluation during laser printing of 3T3 mouse fibroblasts. Biofabrication 2017. [DOI: 10.1088/1758-5090/aa6ed9/.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
15
|
Zhang Z, Chai W, Xiong R, Zhou L, Huang Y. Printing-induced cell injury evaluation during laser printing of 3T3 mouse fibroblasts. Biofabrication 2017. [PMID: 28631624 DOI: 10.1088/1758-5090/aa6ed9/] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Three-dimensional bioprinting has emerged as a promising solution for the freeform fabrication of living cellular constructs, which can be used for tissue/organ transplantation and tissue models. During bioprinting, some living cells are unavoidably injured and may become necrotic or apoptotic cells. This study aims to investigate the printing-induced cell injury and evaluates injury types of post-printing cells using the annexin V/7-aminoactinomycin D and FAM-DEVD-FMK/propidium iodide assays during laser printing of NIH 3T3 mouse fibroblasts. As observed, the percentage of post-printing early apoptotic mouse fibroblasts increases with the incubation time, indicating that post-printing apoptotic mouse fibroblasts have different initiation lag times of apoptosis due to different levels of mechanical stress exerted during laser printing. Post-printing necrotic mouse fibroblasts can be detected immediately after printing, while post-printing early apoptotic mouse fibroblasts need time to develop into a late apoptotic stage. The minimum time needed for post-printing early apoptotic mouse fibroblasts to complete their apoptosis pathway and transition into late apoptotic mouse fibroblasts is from 4 h to 5 h post-printing. The resulting knowledge of the evolution of different apoptotic post-printing mouse fibroblasts will help better design future experiments to quantitatively determine, model, and mitigate the post-printing cell injury based on molecular signal pathway modeling.
Collapse
Affiliation(s)
- Zhengyi Zhang
- School of Naval Architecture and Ocean Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China. Dept. of Mechanical and Aerospace Engineering, Univ. of Florida, Gainesville, FL 32611, United States of America
| | | | | | | | | |
Collapse
|
16
|
Guo FX, Hu YW, Zheng L, Wang Q. Shear Stress in Autophagy and Its Possible Mechanisms in the Process of Atherosclerosis. DNA Cell Biol 2017; 36:335-346. [PMID: 28287831 DOI: 10.1089/dna.2017.3649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy can eliminate harmful components and maintain cellular homeostasis in response to a series of extracellular insults in eukaryotes. More and more studies show that autophagy plays vital roles in the development of atherosclerosis. Atherosclerosis is a multifactorial disease and shear stress acts as a key role in its process. Understanding the role of shear stress in autophagy may offer insight into atherosclerosis therapies, especially emerging targeted therapy. In this article, we retrospect related studies to summarize the present comprehension of the association between autophagy and atherosclerosis onset and progression.
Collapse
Affiliation(s)
- Feng-Xia Guo
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| |
Collapse
|
17
|
Wang Y, Qiu J, Luo S, Xie X, Zheng Y, Zhang K, Ye Z, Liu W, Gregersen H, Wang G. High shear stress induces atherosclerotic vulnerable plaque formation through angiogenesis. Regen Biomater 2016; 3:257-67. [PMID: 27482467 PMCID: PMC4966293 DOI: 10.1093/rb/rbw021] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022] Open
Abstract
Rupture of atherosclerotic plaques causing thrombosis is the main cause of acute coronary syndrome and ischemic strokes. Inhibition of thrombosis is one of the important tasks developing biomedical materials such as intravascular stents and vascular grafts. Shear stress (SS) influences the formation and development of atherosclerosis. The current review focuses on the vulnerable plaques observed in the high shear stress (HSS) regions, which localizes at the proximal region of the plaque intruding into the lumen. The vascular outward remodelling occurs in the HSS region for vascular compensation and that angiogenesis is a critical factor for HSS which induces atherosclerotic vulnerable plaque formation. These results greatly challenge the established belief that low shear stress is important for expansive remodelling, which provides a new perspective for preventing the transition of stable plaques to high-risk atherosclerotic lesions.
Collapse
Affiliation(s)
- Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Shisui Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Xiang Xie
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Yiming Zheng
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Kang Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Wanqian Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Hans Gregersen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China; Taiji Group Co, Ltd, Chongqing, 401147, China
| |
Collapse
|
18
|
Huang CH, Ciou JS, Chen ST, Kok VC, Chung Y, Tsai JJP, Kurubanjerdjit N, Huang CYF, Ng KL. Identify potential drugs for cardiovascular diseases caused by stress-induced genes in vascular smooth muscle cells. PeerJ 2016; 4:e2478. [PMID: 27703845 PMCID: PMC5045879 DOI: 10.7717/peerj.2478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/23/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Abnormal proliferation of vascular smooth muscle cells (VSMC) is a major cause of cardiovascular diseases (CVDs). Many studies suggest that vascular injury triggers VSMC dedifferentiation, which results in VSMC changes from a contractile to a synthetic phenotype; however, the underlying molecular mechanisms are still unclear. METHODS In this study, we examined how VSMC responds under mechanical stress by using time-course microarray data. A three-phase study was proposed to investigate the stress-induced differentially expressed genes (DEGs) in VSMC. First, DEGs were identified by using the moderated t-statistics test. Second, more DEGs were inferred by using the Gaussian Graphical Model (GGM). Finally, the topological parameters-based method and cluster analysis approach were employed to predict the last batch of DEGs. To identify the potential drugs for vascular diseases involve VSMC proliferation, the drug-gene interaction database, Connectivity Map (cMap) was employed. Success of the predictions were determined using in-vitro data, i.e. MTT and clonogenic assay. RESULTS Based on the differential expression calculation, at least 23 DEGs were found, and the findings were qualified by previous studies on VSMC. The results of gene set enrichment analysis indicated that the most often found enriched biological processes are cell-cycle-related processes. Furthermore, more stress-induced genes, well supported by literature, were found by applying graph theory to the gene association network (GAN). Finally, we showed that by processing the cMap input queries with a cluster algorithm, we achieved a substantial increase in the number of potential drugs with experimental IC50 measurements. With this novel approach, we have not only successfully identified the DEGs, but also improved the DEGs prediction by performing the topological and cluster analysis. Moreover, the findings are remarkably validated and in line with the literature. Furthermore, the cMap and DrugBank resources were used to identify potential drugs and targeted genes for vascular diseases involve VSMC proliferation. Our findings are supported by in-vitro experimental IC50, binding activity data and clinical trials. CONCLUSION This study provides a systematic strategy to discover potential drugs and target genes, by which we hope to shed light on the treatments of VSMC proliferation associated diseases.
Collapse
Affiliation(s)
- Chien-Hung Huang
- Department of Computer Science and Information Engineering, National Formosa University, Yun-Lin, Taiwan
| | - Jin-Shuei Ciou
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Shun-Tsung Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Victor C. Kok
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Division of Medical Oncology, Kuang Tien General Hospital Cancer Center, Taichung, Taiwan
| | - Yi Chung
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jeffrey J. P. Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ka-Lok Ng
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
19
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 292] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
20
|
Shear enhances thrombopoiesis and formation of microparticles that induce megakaryocytic differentiation of stem cells. Blood 2014; 124:2094-103. [PMID: 24948658 DOI: 10.1182/blood-2014-01-547927] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In vivo visualization of thrombopoiesis suggests an important role for shear flow in platelet biogenesis. In vitro, shear stress was shown to accelerate proplatelet formation from mature megakaryocytes (Mks). Yet, the role of biomechanical forces on Mk biology and platelet biogenesis remains largely unexplored. In this study, we investigated the impact of shear stress on Mk maturation and formation of platelet-like particles (PLPs), pro/preplatelets (PPTs), and Mk microparticles (MkMPs), and furthermore, we explored a physiological role for MkMPs. We found that shear accelerated DNA synthesis of immature Mks in an exposure time- and shear stress level-dependent manner. Both phosphatidylserine exposure and caspase-3 activation were enhanced by shear stress. Exposure to physiological shear dramatically increased generation of PLPs/PPTs and MkMPs by up to 10.8 and 47-fold, respectively. Caspase-3 inhibition reduced shear-induced PLP/PPT and MkMP formation. PLPs generated under shear flow displayed improved functionality as assessed by CD62P exposure and fibrinogen binding. Significantly, coculture of MkMPs with hematopoietic stem and progenitor cells promoted hematopoietic stem and progenitor cell differentiation to mature Mks synthesizing α- and dense-granules, and forming PPTs without exogenous thrombopoietin, thus identifying a novel and unexplored potential physiological role for MkMPs.
Collapse
|
21
|
Modarreszadeh S, Abouali O, Ghaffarieh A, Ahmadi G. Physiology of aqueous humor dynamic in the anterior chamber due to rapid eye movement. Physiol Behav 2014; 135:112-8. [PMID: 24866913 DOI: 10.1016/j.physbeh.2014.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 04/28/2014] [Accepted: 05/16/2014] [Indexed: 11/30/2022]
Abstract
The nature of aqueous humor (AH) mixing in the anterior chamber (AC) of the human eye due to rapid eye movement (REM) has not been fully understood and has been somewhat a controversial issue. This study uses a computational modeling approach to shed light on this issue. For this purpose a numerical method was developed and used to solve the mathematical equations governing the flow and mixing of aqueous humor motion in the eye subjected to such movements. Based on the experimental measurements available in the literature for the average and maximum amplitudes of the eye movements, a harmonic model for the REM was developed. The corresponding instantaneous and time-averaged velocity fields were evaluated. The simulation results showed that, contrary to earlier reports, the REM led to complex flow structures and a 3-D mixing of AH in the anterior chamber. In addition, the mixing velocity increased in direct proportion to the REM amplitudes. Thus, the AC flow generated by REM could carry nutrients to the posterior surface of the cornea during the sleep. Furthermore, the shear stress acting on the corneal endothelial cells due to REM was computed and compared with that of buoyancy driven flow in the AC due to temperature gradient. It was found that the shear stress generated by REM is much higher than that introduced by the natural convection. A video file for providing a better understanding of the AH mixing process in the AC was also prepared. This video is available on the web.
Collapse
Affiliation(s)
| | - Omid Abouali
- School of Mechanical Engineering, Shiraz University, Shiraz, Iran.
| | - Alireza Ghaffarieh
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| | - Goodarz Ahmadi
- Department of Aeronautical and Mechanical Engineering, Clarkson University, Potsdam, NY, USA
| |
Collapse
|
22
|
Tarbell JM, Shi ZD, Dunn J, Jo H. Fluid Mechanics, Arterial Disease, and Gene Expression. ANNUAL REVIEW OF FLUID MECHANICS 2014; 46:591-614. [PMID: 25360054 PMCID: PMC4211638 DOI: 10.1146/annurev-fluid-010313-141309] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
This review places modern research developments in vascular mechanobiology in the context of hemodynamic phenomena in the cardiovascular system and the discrete localization of vascular disease. The modern origins of this field are traced, beginning in the 1960s when associations between flow characteristics, particularly blood flow-induced wall shear stress, and the localization of atherosclerotic plaques were uncovered, and continuing to fluid shear stress effects on the vascular lining endothelial) cells (ECs), including their effects on EC morphology, biochemical production, and gene expression. The earliest single-gene studies and genome-wide analyses are considered. The final section moves from the ECs lining the vessel wall to the smooth muscle cells and fibroblasts within the wall that are fluid me chanically activated by interstitial flow that imposes shear stresses on their surfaces comparable with those of flowing blood on EC surfaces. Interstitial flow stimulates biochemical production and gene expression, much like blood flow on ECs.
Collapse
Affiliation(s)
- John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065
| | - Jessilyn Dunn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322
| |
Collapse
|
23
|
Qiu J, Zheng Y, Hu J, Liao D, Gregersen H, Deng X, Fan Y, Wang G. Biomechanical regulation of vascular smooth muscle cell functions: from in vitro to in vivo understanding. J R Soc Interface 2013; 11:20130852. [PMID: 24152813 DOI: 10.1098/rsif.2013.0852] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) have critical functions in vascular diseases. Haemodynamic factors are important regulators of VSMC functions in vascular pathophysiology. VSMCs are physiologically active in the three-dimensional matrix and interact with the shear stress sensor of endothelial cells (ECs). The purpose of this review is to illustrate how haemodynamic factors regulate VSMC functions under two-dimensional conditions in vitro or three-dimensional co-culture conditions in vivo. Recent advances show that high shear stress induces VSMC apoptosis through endothelial-released nitric oxide and low shear stress upregulates VSMC proliferation and migration through platelet-derived growth factor released by ECs. This differential regulation emphasizes the need to construct more actual environments for future research on vascular diseases (such as atherosclerosis and hypertension) and cardiovascular tissue engineering.
Collapse
Affiliation(s)
- Juhui Qiu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, College of Bioengineering, Chongqing University, , Chongqing 400044, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Van der Heiden K, Gijsen FJH, Narracott A, Hsiao S, Halliday I, Gunn J, Wentzel JJ, Evans PC. The effects of stenting on shear stress: relevance to endothelial injury and repair. Cardiovasc Res 2013; 99:269-75. [PMID: 23592806 DOI: 10.1093/cvr/cvt090] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Stent deployment following balloon angioplasty is used routinely to treat coronary artery disease. These interventions cause damage and loss of endothelial cells (EC), and thus promote in-stent thrombosis and restenosis. Injured arteries are repaired (intrinsically) by locally derived EC and by circulating endothelial progenitor cells which migrate and proliferate to re-populate denuded regions. However, re-endothelialization is not always complete and often dysfunctional. Moreover, the molecular and biomechanical mechanisms that control EC repair and function in stented segments are poorly understood. Here, we propose that stents modify endothelial repair processes, in part, by altering fluid shear stress, a mechanical force that influences EC migration and proliferation. A more detailed understanding of the biomechanical processes that control endothelial healing would provide a platform for the development of novel therapeutic approaches to minimize damage and promote vascular repair in stented arteries.
Collapse
Affiliation(s)
- Kim Van der Heiden
- Biomedical Engineering, Department Cardiology, ErasmusMC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Chan DD, Van Dyke WS, Bahls M, Connell SD, Critser P, Kelleher JE, Kramer MA, Pearce SM, Sharma S, Neu CP. Mechanostasis in apoptosis and medicine. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2011; 106:517-24. [PMID: 21846479 DOI: 10.1016/j.pbiomolbio.2011.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 08/02/2011] [Indexed: 10/17/2022]
Abstract
Mechanostasis describes a complex and dynamic process where cells maintain equilibrium in response to mechanical forces. Normal physiological loading modes and magnitudes contribute to cell proliferation, tissue growth, differentiation and development. However, cell responses to abnormal forces include compensatory apoptotic mechanisms that may contribute to the development of tissue disease and pathological conditions. Mechanotransduction mechanisms tightly regulate the cell response through discrete signaling pathways. Here, we provide an overview of links between pro- and anti-apoptotic signaling and mechanotransduction signaling pathways, and identify potential clinical applications for treatments of disease by exploiting mechanically-linked apoptotic pathways.
Collapse
Affiliation(s)
- D D Chan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fluid flow mechanotransduction in vascular smooth muscle cells and fibroblasts. Ann Biomed Eng 2011; 39:1608-19. [PMID: 21479754 DOI: 10.1007/s10439-011-0309-2] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/04/2011] [Indexed: 12/29/2022]
Abstract
Understanding how vascular wall endothelial cells (ECs), smooth muscle cells (SMCs), and fibroblasts (FBs) sense and transduce the stimuli of hemodynamic forces (shear stress, cyclic strain, and hydrostatic pressure) into intracellular biochemical signals is critical to prevent vascular disease development and progression. ECs lining the vessel lumen directly sense alterations in blood flow shear stress and then communicate with medial SMCs and adventitial FBs to regulate vessel function and disease. Shear stress mechanotransduction in ECs has been extensively studied and reviewed. In the case of endothelial damage, blood flow shear stress may directly act on the superficial layer of SMCs and transmural interstitial flow may be elevated on medial SMCs and adventitial FBs. Therefore, it is also important to investigate direct shear effects on vascular SMCs as well as FBs. The work published in the last two decades has shown that shear stress and interstitial flow have significant influences on vascular SMCs and FBs. This review summarizes work that considered direct shear effects on SMCs and FBs and provides the first comprehensive overview of the underlying mechanisms that modulate SMC secretion, alignment, contraction, proliferation, apoptosis, differentiation, and migration in response to 2-dimensional (2D) laminar, pulsatile, and oscillating flow shear stresses and 3D interstitial flow. A mechanistic model of flow sensing by SMCs is also provided to elucidate possible mechanotransduction pathways through surface glycocalyx, integrins, membrane receptors, ion channels, and primary cilia. Understanding flow-mediated mechanotransduction in SMCs and FBs and the interplay with ECs should be helpful in exploring strategies to prevent flow-initiated atherosclerosis and neointima formation and has implications in vascular tissue engineering.
Collapse
|
27
|
Kang H, Fan Y, Deng X. Vascular smooth muscle cell glycocalyx modulates shear-induced proliferation, migration, and NO production responses. Am J Physiol Heart Circ Physiol 2010; 300:H76-83. [PMID: 21037235 DOI: 10.1152/ajpheart.00905.2010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The endothelial cell glycocalyx, a structure coating the luminal surface of the vascular endothelium, and its related mechanotransduction have been studied by many over the last decade. However, the role of vascular smooth muscle cells (SMCs) glycocalyx in cell mechanotransduction has triggered little attention. This study addressed the role of heparan sulfate proteoglycans (HSPGs), a major component of the glycocalyx, in the shear-induced proliferation, migration, and nitric oxide (NO) production of the rat aortic smooth muscle cells (RASMCs). A parallel plate flow chamber and a peristaltic pump were employed to expose RASMC monolayers to a physiological level of shear stress (12 dyn/cm(2)). Heparinase III (Hep.III) was applied to selectively degrade heparan sulfate on the SMC surface. Cell proliferation, migration, and NO production rates were determined and compared among the following four groups of cells: 1) untreated with no flow, 2) Hep.III treatment with no flow, 3) untreated with flow of 12 dyn/cm(2) exposure, and 4) Hep.III treatment with flow of 12 dyn/cm(2) exposure. It was observed that flow-induced shear stress significantly suppressed SMC proliferation and migration, whereas cells preferred to aligning along the direction of flow and NO production were enhanced substantially. However, those responses were not found in the cells with Hep.III treatment. Under flow condition, the heparinase III-treated cells remained randomly oriented and proliferated as if there were no flow presence. Disruption of HSPG also enhanced wound closure and inhibited shear-induced NO production significantly. This study suggests that HSPG may play a pivotal role in mechanotransduction of SMCs.
Collapse
Affiliation(s)
- Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science & Medical Engineering, Beihang University, Beijing, China
| | | | | |
Collapse
|
28
|
Shi ZD, Abraham G, Tarbell JM. Shear stress modulation of smooth muscle cell marker genes in 2-D and 3-D depends on mechanotransduction by heparan sulfate proteoglycans and ERK1/2. PLoS One 2010; 5:e12196. [PMID: 20808940 PMCID: PMC2922372 DOI: 10.1371/journal.pone.0012196] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 07/25/2010] [Indexed: 01/31/2023] Open
Abstract
Background During vascular injury, vascular smooth muscle cells (SMCs) and fibroblasts/myofibroblasts (FBs/MFBs) are exposed to altered luminal blood flow or transmural interstitial flow. We investigate the effects of these two types of fluid flows on the phenotypes of SMCs and MFBs and the underlying mechanotransduction mechanisms. Methodology/Principal Findings Exposure to 8 dyn/cm2 laminar flow shear stress (2-dimensional, 2-D) for 15 h significantly reduced expression of α-smooth muscle actin (α-SMA), smooth muscle protein 22 (SM22), SM myosin heavy chain (SM-MHC), smoothelin, and calponin. Cells suspended in collagen gels were exposed to interstitial flow (1 cmH2O, ∼0.05 dyn/cm2, 3-D), and after 6 h of exposure, expression of SM-MHC, smoothelin, and calponin were significantly reduced, while expression of α-SMA and SM22 were markedly enhanced. PD98059 (an ERK1/2 inhibitor) and heparinase III (an enzyme to cleave heparan sulfate) significantly blocked the effects of laminar flow on gene expression, and also reversed the effects of interstitial flow on SM-MHC, smoothelin, and calponin, but enhanced interstitial flow-induced expression of α-SMA and SM22. SMCs and MFBs have similar responses to fluid flow. Silencing ERK1/2 completely blocked the effects of both laminar flow and interstitial flow on SMC marker gene expression. Western blotting showed that both types of flows induced ERK1/2 activation that was inhibited by disruption of heparan sulfate proteoglycans (HSPGs). Conclusions/Significance The results suggest that HSPG-mediated ERK1/2 activation is an important mechanotransduction pathway modulating SMC marker gene expression when SMCs and MFBs are exposed to flow. Fluid flow may be involved in vascular remodeling and lesion formation by affecting phenotypes of vascular wall cells. This study has implications in understanding the flow-related mechanobiology in vascular lesion formation, tumor cell invasion, and stem cell differentiation.
Collapse
Affiliation(s)
- Zhong-Dong Shi
- Department of Biomedical Engineering, The City College of New York, The City University of New York (CUNY), New York, New York, United States of America
- * E-mail: (ZDS); (JMT)
| | - Giya Abraham
- Department of Biomedical Engineering, The City College of New York, The City University of New York (CUNY), New York, New York, United States of America
| | - John M. Tarbell
- Department of Biomedical Engineering, The City College of New York, The City University of New York (CUNY), New York, New York, United States of America
- * E-mail: (ZDS); (JMT)
| |
Collapse
|
29
|
Tissue factor pathway inhibitor-2 is induced by fluid shear stress in vascular smooth muscle cells and affects cell proliferation and survival. J Vasc Surg 2010; 52:167-75. [PMID: 20537494 DOI: 10.1016/j.jvs.2010.02.282] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 02/24/2010] [Accepted: 02/24/2010] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (SMCs) are exposed to fluid shear stress (FSS) after interventional procedures such as balloon-angioplasty. Whereas the effects of hemodynamic forces on endothelial cells are explored in detail, the influence of FSS on smooth muscle cell function is poorly characterized. Here, we investigated the effect of FSS on SMC gene expression and function. METHODS Laminar FSS of arterial level (14 dynes/cm(2)) was applied to SMC cultures for 24 hours in a parallel-plate flow chamber. The effect of FSS on gene expression was first screened with microarray technology, and results further verified by real time polymerase chain reaction (RT-PCR) and immunoblotting. Tissue factor pathway inhibitor-2 (TFPI-2) and caspase-3 protein expression was studied in the rat carotid artery after balloon-injury, and the effect of TFPI-2 on SMC DNA synthesis and apoptosis was examined in vitro. RESULTS Microarrays identified TFPI-2 as one of the most differentially expressed gene by FSS in cultured SMCs (P < .001). Gene set enrichment analysis revealed significant regulation of genes linked to proliferation, apoptosis, and cell cycle regulation. TFPI-2 induction was confirmed by RT-PCR and immunoblotting demonstrating a more than 400-fold (P < .001) increase in TFPI-2 mRNA in SMCs exposed to FSS compared with static controls, and a consistent protein upregulation. Functionally, SMC proliferation was decreased by FSS (P < .001), and recombinant TFPI-2 was found to inhibit SMC proliferation (P < .001) and induce SMC apoptosis as indicated by activation of caspase-3 (P < .01). In vivo, TFPI-2 expression was found to be upregulated 5, 10, and 20 hours (P < .01) after rat carotid balloon injury, and immunohistochemistry demonstrated TFPI-2 protein in FSS-exposed luminal SMCs, co-localized with caspase-3 in the rat carotid neointima. CONCLUSION FSS influenced gene expression associated with cell growth and apoptosis in cultured SMCs and strongly induced expression of TFPI-2 mRNA and protein. TFPI-2 was expressed in luminal, FSS-exposed SMCs together with caspase-3 in the rat carotid neointima after balloon injury. Functionally, TFPI-2 may play a role in vessel wall repair by regulating SMC proliferation and survival. Further studies are needed to elucidate the mechanisms by which TFPI-2 controls SMC function.
Collapse
|
30
|
Fitzgerald TN, Muto A, Fancher TT, Brown PB, Martin KA, Muhs BE, Rothman DL, Constable RT, Sampath S, Dardik A. Surgically implantable magnetic resonance angiography coils improve resolution to allow visualization of blood flow dynamics. Ann Vasc Surg 2009; 24:242-53. [PMID: 20036497 DOI: 10.1016/j.avsg.2009.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2009] [Revised: 08/28/2009] [Accepted: 10/23/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Magnetic resonance angiography (MRA) is clinically useful but of limited applicability to small animal models due to poor signal resolution, with typical voxel sizes of 1 mm(3) that are insufficient to analyze vessels of diameter <1 mm. We determined whether surgically implantable, extravascular MRA coils increase signal resolution adequately to examine blood flow dynamics METHODS A custom MRA coil was surgically implanted near the carotid artery of a New Zealand White rabbit. A stenosis was created in the carotid artery to induce complicated, non-laminar flow. Phase contrast images were obtained on multiple axial planes with 3T MRA and through-plane velocity profiles were calculated under laminar and complicated flow conditions. These velocity profiles were fit to a laminar flow model using ordinary least squares in order to quantify the degree of flow complication (Matlab). Flow was also measured with a Doppler flow probe; vessel diameters and flow velocities were compared with duplex ultrasound RESULTS Carotid artery blood flow was 24.7 +/- 2.6 ml/min prior to stenosis creation and reduced to 12.0 +/- 1.7 ml/min following injury (n=3). An MRA voxel size of 0.1 x 0.1 x 5 mm was achieved. The control carotid artery diameter was 1.9 +/- 0.1 mm, and cross-sectional images containing 318 +/- 22 voxels were acquired (n=26). Velocity profiles resembled laminar flow proximal to the stenosis, and then became more complicated just proximal and distal to the stenosis. Laminar flow conditions returned downstream of the stenosis CONCLUSION Implantable, extra-vascular coils enable small MRA voxel sizes to reproducibly calculate complex velocity profiles under both laminar and complicated flow in a small animal model. This technique may be applied to study blood flow dynamics of vessel remodeling and atherogenesis.
Collapse
Affiliation(s)
- Tamara N Fitzgerald
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Shi ZD, Ji XY, Qazi H, Tarbell JM. Interstitial flow promotes vascular fibroblast, myofibroblast, and smooth muscle cell motility in 3-D collagen I via upregulation of MMP-1. Am J Physiol Heart Circ Physiol 2009; 297:H1225-34. [PMID: 19465549 DOI: 10.1152/ajpheart.00369.2009] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neointima formation often occurs in regions where the endothelium has been damaged and the transmural interstitial flow is elevated. Vascular smooth muscle cells (SMCs) and fibroblasts/myofibroblasts (FBs/MFBs) contribute to intimal thickening by migrating from the media and adventitia into the site of injury. In this study, for the first time, the direct effects of interstitial flow on SMC and FB/MFB migration were investigated in an in vitro three-dimensional system. Collagen I gels were used to mimic three-dimensional extracellular matrix (ECM) for rat aortic SMCs and FBs/MFBs. Exposure to interstitial flow induced by 1 cmH(2)O pressure differential (shear stress, approximately 0.05 dyn/cm(2); flow velocity, approximately 0.5 microm/s; and Darcy permeability, approximately 10(-11) cm(2)) substantially enhanced cell motility. Matrix metalloproteinase (MMP) inhibitor (GM-6001) abolished flow-induced migration augmentation, which suggested that the enhanced motility was MMP dependent. The upregulation of MMP-1 played a critical role for the flow-enhanced motility, which was further confirmed by silencing MMP-1 gene expression. Longer exposures to higher flows suppressed the number of migrated cells, although MMP-1 gene expression remained high. This suppression was a result of both flow-induced tissue inhibitor of metalloproteinase-1 upregulation and increased apoptotic and necrotic cell death. Interstitial flow did not affect MMP-2 gene expression or activity in the collagen I gel for any cell type. Our findings shed light on the mechanism by which vascular SMCs and FBs/MFBs contribute to intimal thickening in regions of vascular injury where interstitial flow is elevated.
Collapse
Affiliation(s)
- Zhong-Dong Shi
- Department of Biomedical Engineering, City College of New York, City University of New York, New York 10031, USA
| | | | | | | |
Collapse
|