1
|
Chandurkar MK, Yang M, Rostami M, Han SJ. TRPV4 Dominates High Shear-Induced Initial Traction Response and Long-Term Relaxation Over Piezo1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637570. [PMID: 39990362 PMCID: PMC11844455 DOI: 10.1101/2025.02.10.637570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Modulation of endothelial traction is critical for the responses of endothelial cells to fluid shear stress (FSS), which has profound implications for vascular health and atherosclerosis. Previously, we demonstrated that under high FSS, endothelial cells rapidly increase traction forces, followed by relaxation, with traction aligning in the flow direction. In contrast, low shear preconditioning induces a modest short-term increase in traction (<30 min), followed by a secondary long-term (>14 hr) rise, with traction/cells aligning perpendicular to the flow. The upstream mechanosensors driving these responses, however, remain unknown. Here, we sought the roles of Piezo1 and TRPV4 ion channels in shear-induced traction modulation. We report that HUVECs with Piezo1 silencing reduced the initial traction rise in half under high FSS compared to those by WT cells, while not affecting the traction modulation in response to low FSS or traction/cell alignment to the flow direction. Conversely, cells with siTRPV4 fully abrogated the initial traction rise, as well as alignment of traction and cells, in response to both high and low FSS conditions. Dual inhibition of Piezo1 and TRPV4 further impaired both initial and long-term traction under high FSS. Interestingly, dual-inhibited cells displayed larger initial traction responses to low FSS compared to control cells, suggesting the involvement of alternative calcium-independent pathways that become dominant when both ion channels are nonfunctional. Additionally, either ion channel inhibition led to secondary long-term traction increase even under high FSS condition. These findings suggest that while both Piezo1 and TRPV4 channels contribute to shear mechanotransduction, TRPV4 plays more dominant role than Piezo1 in mediating the initial traction rise and sustaining long-term relaxation under high or low shear stress, highlighting their critical and distinct contributions to endothelial mechanotransduction and remodeling.
Collapse
Affiliation(s)
- Mohanish K Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Manli Yang
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, United States
| | - Majid Rostami
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Sangyoon J Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, United States
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, Michigan, United States
| |
Collapse
|
2
|
Guo P, Chen L, Yang D, Zhang L, Shu C, Li H, Zhu J, Zhou J, Li X. Predictive value of plasma ephrinB2 levels for amputation risk following endovascular revascularization in peripheral artery disease. PeerJ 2024; 12:e17531. [PMID: 38854794 PMCID: PMC11162178 DOI: 10.7717/peerj.17531] [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: 11/06/2023] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Background The aim of this study is to investigate the expression levels of ephrinB2 in patients with lower extremity peripheral arterial disease (PAD) and explore its association with the severity of the disease and the risk of amputation after endovascular revascularization. Methods During the period from March 2021 to March 2023, this study collected blood samples and clinical data from 133 patients diagnosed with lower extremity PAD and 51 healthy volunteer donors. The severity of lower extremity PAD patients was classified using the Rutherford categories. The expression of ephrin-B2 in plasma samples was detected using the Western Blotting. Results Compared to the control group, the levels of serum ephrinB2 in patients were significantly elevated (p < 0.001). Moreover, the plasma EphrinB2 levels were positively correlated with white blood cell counts (r = 0.204, p = 0.018), neutrophil counts (r = 0.174, p = 0.045), and neutrophil-to-lymphocyte ratio (NLR) (r = 0.223, p = 0.009). Furthermore, the AUCs of plasma ephrinB2 level, NLR, and their combination as predictors for amputation events within 30 months after lower extremity PAD endovascular revascularization were 0.659, 0.730 and 0.811. In the high-ephrinB2 group, the incidence of amputation events within 30 months after endovascular revascularization was higher. Conclusions Plasma EphrinB2 levels may be linked to lower extremity PAD development, inflammation, and postoperative amputation. Combining EphrinB2 and NLR can improve amputation prediction accuracy after endovascular revascularization in lower extremity PAD patients.
Collapse
Affiliation(s)
- Pengcheng Guo
- Vascular Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
- Institute of Vascular Diseases, Central South University, Chang Sha, Hunan, China
| | - Lei Chen
- Pharmacy Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
| | - Dafeng Yang
- Cardiology Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
| | - Lei Zhang
- Vascular Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
- Institute of Vascular Diseases, Central South University, Chang Sha, Hunan, China
| | - Chang Shu
- Vascular Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
- Institute of Vascular Diseases, Central South University, Chang Sha, Hunan, China
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huande Li
- Pharmacy Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
| | - Jieting Zhu
- Vascular Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
- Institute of Vascular Diseases, Central South University, Chang Sha, Hunan, China
| | - Jienan Zhou
- Vascular Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
- Institute of Vascular Diseases, Central South University, Chang Sha, Hunan, China
| | - Xin Li
- Vascular Surgery Department, the Secondary Xiangya Hospital, Central South University, Hunan, China
- Institute of Vascular Diseases, Central South University, Chang Sha, Hunan, China
| |
Collapse
|
3
|
Chandurkar MK, Mittal N, Royer-Weeden SP, Lehmann SD, Michels EB, Haarman SE, Severance SA, Rho Y, Han SJ. Transient low shear-stress preconditioning influences long-term endothelial traction and alignment under high shear flow. Am J Physiol Heart Circ Physiol 2024; 326:H1180-H1192. [PMID: 38457352 PMCID: PMC11649189 DOI: 10.1152/ajpheart.00067.2024] [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: 02/02/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Endothelial cells (ECs) within the vascular system encounter fluid shear stress (FSS). High, laminar FSS promotes vasodilation and anti-inflammatory responses, whereas low or disturbed FSS induces dysfunction and inflammation. However, the adaptation of endothelial cells (ECs) to dynamically changing FSS patterns remains underexplored. Here, by combining traction force microscopy with a custom flow chamber, we examined human umbilical vein endothelial cells adapting their traction during transitions from short-term low shear to long-term high shear stress. We discovered that the initial low FSS elevates the traction by only half of the amount in response to direct high FSS even after flow changes to high FSS. However, in the long term under high FSS, the flow started with low FSS triggers a substantial second rise in traction for over 10 h. In contrast, the flow started directly with high FSS results in a quick traction surge followed by a huge reduction below the baseline traction in <30 min. Importantly, we find that the orientation of traction vectors is steered by initial shear exposure. Using Granger causality analysis, we show that the traction that aligns in the flow direction under direct high FSS functionally causes cell alignment toward the flow direction. However, EC traction that orients perpendicular to the flow that starts with temporary low FSS functionally causes cell orientation perpendicular to the flow. Taken together, our findings elucidate the significant influence of initial short-term low FSS on lasting changes in endothelial traction that induces EC alignment.NEW & NOTEWORTHY In our study, we uncover that preconditioning with low shear stress yields enduring impacts on endothelial cell traction and orientation, persisting even after transitioning to high-shear conditions. Using Granger causality analysis, we demonstrate a functional link between the direction of cell traction and subsequent cellular alignment across varying shear environments.
Collapse
Affiliation(s)
- Mohanish K Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Shaina P Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Steven D Lehmann
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Etienne B Michels
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Samuel E Haarman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Scott A Severance
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Yeonwoo Rho
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States
| | - Sangyoon J Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, Michigan, United States
| |
Collapse
|
4
|
Chandurkar MK, Mittal N, Royer-Weeden SP, Lehmann SD, Rho Y, Han SJ. Low Shear in Short-Term Impacts Endothelial Cell Traction and Alignment in Long-Term. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.20.558732. [PMID: 37790318 PMCID: PMC10542130 DOI: 10.1101/2023.09.20.558732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Within the vascular system, endothelial cells (ECs) are exposed to fluid shear stress (FSS), a mechanical force exerted by blood flow that is critical for regulating cellular tension and maintaining vascular homeostasis. The way ECs react to FSS varies significantly; while high, laminar FSS supports vasodilation and suppresses inflammation, low or disturbed FSS can lead to endothelial dysfunction and increase the risk of cardiovascular diseases. Yet, the adaptation of ECs to dynamically varying FSS remains poorly understood. This study focuses on the dynamic responses of ECs to brief periods of low FSS, examining its impact on endothelial traction-a measure of cellular tension that plays a crucial role in how endothelial cells respond to mechanical stimuli. By integrating traction force microscopy (TFM) with a custom-built flow chamber, we analyzed how human umbilical vein endothelial cells (HUVECs) adjust their traction in response to shifts from low to high shear stress. We discovered that initial exposure to low FSS prompts a marked increase in traction force, which continues to rise over 10 hours before slowly decreasing. In contrast, immediate exposure to high FSS causes a quick spike in traction followed by a swift reduction, revealing distinct patterns of traction behavior under different shear conditions. Importantly, the direction of traction forces and the resulting cellular alignment under these conditions indicate that the initial shear experience dictates long-term endothelial behavior. Our findings shed light on the critical influence of short-lived low-shear stress experiences in shaping endothelial function, indicating that early exposure to low FSS results in enduring changes in endothelial contractility and alignment, with significant consequences for vascular health and the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Mohanish K. Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Shaina P. Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Steven D. Lehmann
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Yeonwoo Rho
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI 49931
| | - Sangyoon J. Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, MI 49931
| |
Collapse
|
5
|
Suarez Rodriguez F, Sanlidag S, Sahlgren C. Mechanical regulation of the Notch signaling pathway. Curr Opin Cell Biol 2023; 85:102244. [PMID: 37783031 DOI: 10.1016/j.ceb.2023.102244] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/13/2023] [Accepted: 09/03/2023] [Indexed: 10/04/2023]
Abstract
The mechanical regulation of Notch signaling is an emerging area of interest in cell biology. Notch is essential in many physiological processes in which mechanical stress plays an important role. This review provides an overview of the mechanoregulation of Notch signaling in multiple steps of the pathway. First, we discuss the current knowledge on the direct mechanoregulation of Notch receptor maturation and localization to the membrane and the effect of mechanical stress on the Notch components. Next, we explore how ligand-receptor interactions and membrane dynamics are possible subjects to mechano-regulation, emphasizing the role of cytoskeletal interactions, membrane stiffness, and endocytic complex formation. We further delve into the necessity of tension generation for negative regulatory region (NRR) domain unfolding, facilitated by ligand endocytosis and other microforces. Additionally, we examine the indirect mechano-regulation of S2 and S3 cleavages. Finally, we discuss the mechanoregulation of the Notch intracellular domain (NICD) trafficking and nuclear entry and the impact of mechanical stress on heterochromatin dynamics and nuclear NICD interactions. This review aims to draw attention to the intricate interplay between mechanical cues and Notch signaling regulation, offering novel insights into the multifaceted nature of cellular mechanobiology.
Collapse
Affiliation(s)
- Freddy Suarez Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, FI-20520, Turku, Finland; Turku Bioscience, Åbo Akademi University and University of Turku, Tykistökatu 6, FI-20520, Turku, Finland; InFLAMES Research Flagship Center, Åbo Akademi University and University of Turku, Turku, Finland
| | - Sami Sanlidag
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, FI-20520, Turku, Finland; Turku Bioscience, Åbo Akademi University and University of Turku, Tykistökatu 6, FI-20520, Turku, Finland; InFLAMES Research Flagship Center, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, FI-20520, Turku, Finland; Turku Bioscience, Åbo Akademi University and University of Turku, Tykistökatu 6, FI-20520, Turku, Finland; InFLAMES Research Flagship Center, Åbo Akademi University and University of Turku, Turku, Finland; Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, the Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Ceres, Building Number 7, De Zaale, 5612 AJ, Eindhoven, the Netherlands.
| |
Collapse
|
6
|
Mannion AJ, Holmgren L. Nuclear mechanosensing of the aortic endothelium in health and disease. Dis Model Mech 2023; 16:dmm050361. [PMID: 37909406 PMCID: PMC10629673 DOI: 10.1242/dmm.050361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
The endothelium, the monolayer of endothelial cells that line blood vessels, is exposed to a number of mechanical forces, including frictional shear flow, pulsatile stretching and changes in stiffness influenced by extracellular matrix composition. These forces are sensed by mechanosensors that facilitate their transduction to drive appropriate adaptation of the endothelium to maintain vascular homeostasis. In the aorta, the unique architecture of the vessel gives rise to changes in the fluid dynamics, which, in turn, shape cellular morphology, nuclear architecture, chromatin dynamics and gene regulation. In this Review, we discuss recent work focusing on how differential mechanical forces exerted on endothelial cells are sensed and transduced to influence their form and function in giving rise to spatial variation to the endothelium of the aorta. We will also discuss recent developments in understanding how nuclear mechanosensing is implicated in diseases of the aorta.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| | - Lars Holmgren
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| |
Collapse
|
7
|
Aitken C, Mehta V, Schwartz MA, Tzima E. Mechanisms of endothelial flow sensing. NATURE CARDIOVASCULAR RESEARCH 2023; 2:517-529. [PMID: 39195881 DOI: 10.1038/s44161-023-00276-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/14/2023] [Indexed: 08/29/2024]
Abstract
Fluid shear stress plays a key role in sculpting blood vessels during development, in adult vascular homeostasis and in vascular pathologies. During evolution, endothelial cells evolved several mechanosensors that convert physical forces into biochemical signals, a process termed mechanotransduction. This Review discusses our understanding of endothelial flow sensing and suggests important questions for future investigation.
Collapse
Affiliation(s)
- Claire Aitken
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vedanta Mehta
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - Ellie Tzima
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
8
|
Choi D, Park E, Yu RP, Cooper MN, Cho IT, Choi J, Yu J, Zhao L, Yum JEI, Yu JS, Nakashima B, Lee S, Seong YJ, Jiao W, Koh CJ, Baluk P, McDonald DM, Saraswathy S, Lee JY, Jeon NL, Zhang Z, Huang AS, Zhou B, Wong AK, Hong YK. Piezo1-Regulated Mechanotransduction Controls Flow-Activated Lymphatic Expansion. Circ Res 2022; 131:e2-e21. [PMID: 35701867 PMCID: PMC9308715 DOI: 10.1161/circresaha.121.320565] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Mutations in PIEZO1 (Piezo type mechanosensitive ion channel component 1) cause human lymphatic malformations. We have previously uncovered an ORAI1 (ORAI calcium release-activated calcium modulator 1)-mediated mechanotransduction pathway that triggers lymphatic sprouting through Notch downregulation in response to fluid flow. However, the identity of its upstream mechanosensor remains unknown. This study aimed to identify and characterize the molecular sensor that translates the flow-mediated external signal to the Orai1-regulated lymphatic expansion. METHODS Various mutant mouse models, cellular, biochemical, and molecular biology tools, and a mouse tail lymphedema model were employed to elucidate the role of Piezo1 in flow-induced lymphatic growth and regeneration. RESULTS Piezo1 was found to be abundantly expressed in lymphatic endothelial cells. Piezo1 knockdown in cultured lymphatic endothelial cells inhibited the laminar flow-induced calcium influx and abrogated the flow-mediated regulation of the Orai1 downstream genes, such as KLF2 (Krüppel-like factor 2), DTX1 (Deltex E3 ubiquitin ligase 1), DTX3L (Deltex E3 ubiquitin ligase 3L,) and NOTCH1 (Notch receptor 1), which are involved in lymphatic sprouting. Conversely, stimulation of Piezo1 activated the Orai1-regulated mechanotransduction in the absence of fluid flow. Piezo1-mediated mechanotransduction was significantly blocked by Orai1 inhibition, establishing the epistatic relationship between Piezo1 and Orai1. Lymphatic-specific conditional Piezo1 knockout largely phenocopied sprouting defects shown in Orai1- or Klf2- knockout lymphatics during embryo development. Postnatal deletion of Piezo1 induced lymphatic regression in adults. Ectopic Dtx3L expression rescued the lymphatic defects caused by Piezo1 knockout, affirming that the Piezo1 promotes lymphatic sprouting through Notch downregulation. Consistently, transgenic Piezo1 expression or pharmacological Piezo1 activation enhanced lymphatic sprouting. Finally, we assessed a potential therapeutic value of Piezo1 activation in lymphatic regeneration and found that a Piezo1 agonist, Yoda1, effectively suppressed postsurgical lymphedema development. CONCLUSIONS Piezo1 is an upstream mechanosensor for the lymphatic mechanotransduction pathway and regulates lymphatic growth in response to external physical stimuli. Piezo1 activation presents a novel therapeutic opportunity for preventing postsurgical lymphedema. The Piezo1-regulated lymphangiogenesis mechanism offers a molecular basis for Piezo1-associated lymphatic malformation in humans.
Collapse
Affiliation(s)
- Dongwon Choi
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Eunkyung Park
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Roy P. Yu
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael N. Cooper
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Il-Taeg Cho
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Joshua Choi
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - James Yu
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Luping Zhao
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ji-Eun Irene Yum
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jin Suh Yu
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Brandon Nakashima
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sunju Lee
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Young Jin Seong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wan Jiao
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Chester J. Koh
- Division of Pediatric Urology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Baluk
- Cardiovascular Research Institute, UCSF Helen Diller Family Comprehensive Cancer Center, and Department of Anatomy, University of California, San Francisco, San Francisco, California, USA
| | - Donald M. McDonald
- Cardiovascular Research Institute, UCSF Helen Diller Family Comprehensive Cancer Center, and Department of Anatomy, University of California, San Francisco, San Francisco, California, USA
| | - Sindhu Saraswathy
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jong Y. Lee
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Noo Li Jeon
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Zhenqian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Alex S. Huang
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Alex K. Wong
- Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Young-Kwon Hong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
9
|
Fan L, Liu H, Zhu G, Singh S, Yu Z, Wang S, Luo H, Liu S, Xu Y, Ge J, Jiang D, Pang J. Caspase-4/11 is critical for angiogenesis by repressing Notch1 signaling via inhibiting γ-secretase activity. Br J Pharmacol 2022; 179:4809-4828. [PMID: 35737588 DOI: 10.1111/bph.15904] [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: 11/16/2021] [Revised: 05/23/2022] [Accepted: 05/29/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Notch1 activation mediated by γ-secretase is critical for angiogenesis. GeneCards database predicted that Caspase-4 (CASP4, with murine ortholog CASP11) interacts with presenilin-1, the catalytic core of γ-secretase. Therefore, we investigated the role of CASP4/11 in angiogenesis. EXPERIMENTAL APPROACH In vivo, we studied the role of Casp11 in several angiogenesis mouse models using Casp11 wild-type and knockout mice. In vitro, we detected the effects of CASP4 on endothelial functions and Notch signaling by depleting or overexpressing CASP4 in human umbilical vein endothelial cells (HUVECs). The functional domain responsible for the binding of CASP4 and presenilin-1 was detected by mutagenesis and co-immunoprecipitation. KEY RESULTS Casp11 deficiency remarkably impaired adult angiogenesis in ischemic hindlimbs, melanoma xenografts and Matrigel plugs, but not the developmental angiogenesis of retina. Bone marrow transplantation revealed that the pro-angiogenic effect depended on CASP11 derived from non-hematopoietic cells. CASP4 expression was induced by inflammatory factors and CASP4 knockdown decreased cell viability, proliferation, migration and tube formation in HUVECs. Mechanistically, CASP4/11 deficiency increased Notch1 activation in vivo and in vitro, while CASP4 overexpression repressed Notch1 signaling in HUVECs. Moreover, CASP4 knockdown increased γ-secretase activity. γ-Secretase inhibitor DAPT restored the effects of CASP4 siRNA on Notch1 activation and angiogenesis in HUVECs. Notably, the catalytic activity of CASP4/11 was dispensable. Instead, CASP4 directly interacted with presenilin-1 through the caspase recruitment domain (CARD). CONCLUSIONS AND IMPLICATIONS These findings reveal a critical role of CASP4/11 in adult angiogenesis and make this molecule a promising therapeutic target for angiogenesis-related diseases in the future.
Collapse
Affiliation(s)
- Linlin Fan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hao Liu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guofu Zhu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shekhar Singh
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ze Yu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Hong Luo
- Department of Medical Laboratory, College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Shiying Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Dongyang Jiang
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| |
Collapse
|
10
|
The essential role for endothelial cell sprouting in coronary collateral growth. J Mol Cell Cardiol 2022; 165:158-171. [PMID: 35074317 PMCID: PMC8940680 DOI: 10.1016/j.yjmcc.2022.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/11/2022] [Accepted: 01/16/2022] [Indexed: 12/11/2022]
Abstract
RATIONALE Coronary collateral growth is a natural bypass for ischemic heart diseases. It offers tremendous therapeutic benefit, but the process of coronary collateral growth isincompletely understood due to limited preclinical murine models that would enable interrogation of its mechanisms and processes via genetic modification and lineage tracing. Understanding the processes by which coronary collaterals develop can unlock new therapeutic strategies for ischemic heart disease. OBJECTIVE To develop a murine model of coronary collateral growth by repetitive ischemia and investigate whether capillary endothelial cells could contribute to the coronary collateral formation in an adult mouse heart after repetitive ischemia by lineage tracing. METHODS AND RESULTS A murine model of coronary collateral growth was developed using short episodes of repetitive ischemia. Repetitive ischemia stimulation resulted in robust collateral growth in adult mouse hearts, validated by high-resolution micro-computed tomography. Repetitive ischemia-induced collateral formation compensated ischemia caused by occlusion of the left anterior descending artery. Cardiac function improved during ischemia after repetitive ischemia, suggesting the improvement of coronary blood flow. A capillary-specific Cre driver (Apln-CreER) was used for lineage tracing capillary endothelial cells. ROSA mT/mG reporter mice crossed with the Apln-CreER transgene mice underwent a 17 days' repetitive ischemia protocol for coronary collateral growth. Two-photon and confocal microscopy imaging of heart slices revealed repetitive ischemia-induced coronary collateral growth initiated from sprouting Apelin+ endothelial cells. Newly formed capillaries in the collateral-dependent zone expanded in diameter upon repetitive ischemia stimulation and arterialized with smooth muscle cell recruitment, forming mature coronary arteries. Notably, pre-existing coronary arteries and arterioles were not Apelin+, and all Apelin+ collaterals arose from sprouting capillaries. Cxcr4, Vegfr2, Jag1, Mcp1, and Hif1⍺ mRNA levels in the repetitive ischemia-induced hearts were also upregulated at the early stage of coronary collateral growth, suggesting angiogenic signaling pathways are activated for coronary collaterals formation during repetitive ischemia. CONCLUSIONS We developed a murine model of coronary collateral growth induced by repetitive ischemia. Our lineage tracing study shows that sprouting endothelial cells contribute to coronary collateral growth in adult mouse hearts. For the first time, sprouting angiogenesis is shown to give rise to mature coronary arteries in response to repetitive ischemia in the adult mouse hearts.
Collapse
|
11
|
Mehta V, Pang KL, Givens CS, Chen Z, Huang J, Sweet DT, Jo H, Reader JS, Tzima E. Mechanical forces regulate endothelial-to-mesenchymal transition and atherosclerosis via an Alk5-Shc mechanotransduction pathway. SCIENCE ADVANCES 2021; 7:7/28/eabg5060. [PMID: 34244146 PMCID: PMC8270486 DOI: 10.1126/sciadv.abg5060] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/27/2021] [Indexed: 05/06/2023]
Abstract
The response of endothelial cells to mechanical forces is a critical determinant of vascular health. Vascular pathologies, such as atherosclerosis, characterized by abnormal mechanical forces are frequently accompanied by endothelial-to-mesenchymal transition (EndMT). However, how forces affect the mechanotransduction pathways controlling cellular plasticity, inflammation, and, ultimately, vessel pathology is poorly understood. Here, we identify a mechanoreceptor that is sui generis for EndMT and unveil a molecular Alk5-Shc pathway that leads to EndMT and atherosclerosis. Depletion of Alk5 abrogates shear stress-induced EndMT responses, and genetic targeting of endothelial Shc reduces EndMT and atherosclerosis in areas of disturbed flow. Tensional force and reconstitution experiments reveal a mechanosensory function for Alk5 in EndMT signaling that is unique and independent of other mechanosensors. Our findings are of fundamental importance for understanding how mechanical forces regulate biochemical signaling, cell plasticity, and vascular disease.
Collapse
Affiliation(s)
- Vedanta Mehta
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kar-Lai Pang
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christopher S Givens
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhongming Chen
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jianhua Huang
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel T Sweet
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - John S Reader
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ellie Tzima
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Chen D, Schwartz MA, Simons M. Developmental Perspectives on Arterial Fate Specification. Front Cell Dev Biol 2021; 9:691335. [PMID: 34249941 PMCID: PMC8269928 DOI: 10.3389/fcell.2021.691335] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Blood vessel acquisition of arterial or venous fate is an adaptive phenomenon in response to increasing blood circulation during vascular morphogenesis. The past two decades of effort in this field led to development of a widely accepted paradigm of molecular regulators centering on VEGF and Notch signaling. More recent findings focused on shear stress-induced cell cycle arrest as a prerequisite for arterial specification substantially modify this traditional understanding. This review aims to summarize key molecular mechanisms that work in concert to drive the acquisition of arterial fate in two distinct developmental settings of vascular morphogenesis: de novo vasculogenesis of the dorsal aorta and postnatal retinal angiogenesis. We will also discuss the questions and conceptual controversies that potentially point to novel directions of investigation and possible clinical relevance.
Collapse
Affiliation(s)
- Dongying Chen
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
13
|
Bosseboeuf E, Raimondi C. Signalling, Metabolic Pathways and Iron Homeostasis in Endothelial Cells in Health, Atherosclerosis and Alzheimer's Disease. Cells 2020; 9:cells9092055. [PMID: 32911833 PMCID: PMC7564205 DOI: 10.3390/cells9092055] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells drive the formation of new blood vessels in physiological and pathological contexts such as embryonic development, wound healing, cancer and ocular diseases. Once formed, all vessels of the vasculature system present an endothelial monolayer (the endothelium), lining the luminal wall of the vessels, that regulates gas and nutrient exchange between the circulating blood and tissues, contributing to maintaining tissue and vascular homeostasis. To perform their functions, endothelial cells integrate signalling pathways promoted by growth factors, cytokines, extracellular matrix components and signals from mechanosensory complexes sensing the blood flow. New evidence shows that endothelial cells rely on specific metabolic pathways for distinct cellular functions and that the integration of signalling and metabolic pathways regulates endothelial-dependent processes such as angiogenesis and vascular homeostasis. In this review, we provide an overview of endothelial functions and the recent advances in understanding the role of endothelial signalling and metabolism in physiological processes such as angiogenesis and vascular homeostasis and vascular diseases. Also, we focus on the signalling pathways promoted by the transmembrane protein Neuropilin-1 (NRP1) in endothelial cells, its recently discovered role in regulating mitochondrial function and iron homeostasis and the role of mitochondrial dysfunction and iron in atherosclerosis and neurodegenerative diseases.
Collapse
|
14
|
Douglas G, Mehta V, Al Haj Zen A, Akoumianakis I, Goel A, Rashbrook VS, Trelfa L, Donovan L, Drydale E, Chuaiphichai S, Antoniades C, Watkins H, Kyriakou T, Tzima E, Channon KM. A key role for the novel coronary artery disease gene JCAD in atherosclerosis via shear stress mechanotransduction. Cardiovasc Res 2020; 116:1863-1874. [PMID: 31584065 PMCID: PMC7449560 DOI: 10.1093/cvr/cvz263] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/13/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS Genome-wide association studies (GWAS) have consistently identified an association between coronary artery disease (CAD) and a locus on chromosome 10 containing a single gene, JCAD (formerly KIAA1462). However, little is known about the mechanism by which JCAD could influence the development of atherosclerosis. METHODS AND RESULTS Vascular function was quantified in subjects with CAD by flow-mediated dilatation (FMD) and vasorelaxation responses in isolated blood vessel segments. The JCAD risk allele identified by GWAS was associated with reduced FMD and reduced endothelial-dependent relaxations. To study the impact of loss of Jcad on atherosclerosis, Jcad-/- mice were crossed to an ApoE-/- background and fed a high-fat diet from 6 to16 weeks of age. Loss of Jcad did not affect blood pressure or heart rate. However, Jcad-/-ApoE-/- mice developed significantly less atherosclerosis in the aortic root and the inner curvature of the aortic arch. En face analysis revealed a striking reduction in pro-inflammatory adhesion molecules at sites of disturbed flow on the endothelial cell layer of Jcad-/- mice. Loss of Jcad lead to a reduced recovery perfusion in response to hind limb ischaemia, a model of altered in vivo flow. Knock down of JCAD using siRNA in primary human aortic endothelial cells significantly reduced the response to acute onset of flow, as evidenced by reduced phosphorylation of NF-КB, eNOS, and Akt. CONCLUSION The novel CAD gene JCAD promotes atherosclerotic plaque formation via a role in the endothelial cell shear stress mechanotransduction pathway.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/physiopathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/physiopathology
- Aortic Diseases/prevention & control
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/physiopathology
- Atherosclerosis/prevention & control
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cells, Cultured
- Coronary Artery Disease/genetics
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/physiopathology
- Coronary Circulation
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Disease Models, Animal
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Genome-Wide Association Study
- Hindlimb/blood supply
- Humans
- Ischemia/genetics
- Ischemia/metabolism
- Ischemia/physiopathology
- Male
- Mechanotransduction, Cellular
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- NF-kappa B/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Phosphorylation
- Plaque, Atherosclerotic
- Proto-Oncogene Proteins c-akt
- Stress, Mechanical
Collapse
Affiliation(s)
- Gillian Douglas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Vedanta Mehta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ayman Al Haj Zen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar Foundation, Doha, Qatar
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Victoria S Rashbrook
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lucy Trelfa
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lucy Donovan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Edward Drydale
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Surawee Chuaiphichai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Theodosios Kyriakou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ellie Tzima
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
15
|
Ma T, Bai YP. The hydromechanics in arteriogenesis. Aging Med (Milton) 2020; 3:169-177. [PMID: 33103037 PMCID: PMC7574636 DOI: 10.1002/agm2.12101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/23/2020] [Accepted: 02/23/2020] [Indexed: 12/15/2022] Open
Abstract
Coronary heart diseases are tightly associated with aging. Although current revascularization therapies, such as percutaneous coronary interventions (PCI) and coronary artery bypass graft (CABG), improve the clinical outcomes of patients with coronary diseases, their application and therapeutic effects are limited in elderly patients. Thus, developing novel therapeutic strategies, like prompting collateral development or the process of arteriogenesis, is necessary for the treatment of the elderly with coronary diseases. Arteriogenesis (ie, the vascular remodeling from pre‐existent arterioles to collateral conductance networks) functions as an essential compensation for tissue hypoperfusion caused by artery occlusion or stenosis, and its mechanisms remain to be elucidated. In this review, we will summarize the roles of the major hydromechanical components in laminar conditions in arteriogenesis, and discuss the potential effects of disturbed flow components in non‐laminar conditions.
Collapse
Affiliation(s)
- Tianqi Ma
- Department of Geriatric Medicine Xiangya Hospital Central South University Changsha China
| | - Yong-Ping Bai
- Department of Geriatric Medicine Xiangya Hospital Central South University Changsha China
| |
Collapse
|
16
|
Lin XC, Pan M, Zhu LP, Sun Q, Zhou ZS, Li CC, Zhang GG. NFAT5 promotes arteriogenesis via MCP-1-dependent monocyte recruitment. J Cell Mol Med 2019; 24:2052-2063. [PMID: 31883300 PMCID: PMC6991654 DOI: 10.1111/jcmm.14904] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/28/2019] [Accepted: 10/05/2019] [Indexed: 01/24/2023] Open
Abstract
Studies have demonstrated that nuclear factor of activated T cells 5 (NFAT5) is not only a tonicity‐responsive transcription factor but also activated by other stimuli, so we aim to investigate whether NFAT5 participates in collateral arteries formation in rats. We performed femoral artery ligature (FAL) in rats for hindlimb ischaemia model and found that NFAT5 was up‐regulated in rat adductors with FAL compared with sham group. Knockdown of NFAT5 with locally injection of adenovirus‐mediated NFAT5‐shRNA in rats significantly inhibited hindlimb blood perfusion recovery and arteriogenesis. Moreover, NFAT5 knockdown decreased macrophages infiltration and monocyte chemotactic protein‐1 (MCP‐1) expression in rats adductors. In vitro, with interleukin‐1β (IL‐1β) stimulation and loss‐of‐function studies, we demonstrated that NFAT5 knockdown inhibits MCP‐1 expression in endothelial cells and chemotaxis of THP‐1 cells regulated by ERK1/2 pathway. More importantly, exogenous MCP‐1 delivery could recover hindlimb blood perfusion, promote arteriogenesis and macrophages infiltration in rats after FAL, which were depressed by NFAT5 knockdown. Besides, NFAT5 knockdown also inhibited angiogenesis in gastrocnemius muscles in rats. Our results indicate that NFAT5 is a critical regulator of arteriogenesis and angiogenesis via MCP‐1‐dependent monocyte recruitment, suggesting that NFAT5 may represent an alternative therapeutic target for ischaemic diseases.
Collapse
Affiliation(s)
- Xing-Chi Lin
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Miao Pan
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Ping Zhu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Sun
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Shi Zhou
- Department of Laboratory Animal, Xiangya School of Medicine, Central South University, Changsha, China
| | - Chuan-Chang Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Gang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Cardiovascular Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Min E, Schwartz MA. Translocating transcription factors in fluid shear stress-mediated vascular remodeling and disease. Exp Cell Res 2019; 376:92-97. [PMID: 30633880 DOI: 10.1016/j.yexcr.2019.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
Endothelial cells are exposed to fluid shear stress profiles that vary in magnitude, pulsatility, and directionality due to regional variations in blood vessel structure. Laminar flow at physiological levels is atheroprotective; multidirectional or reversing low (disturbed) flow promotes inflammation and disease; and high or low laminar flow promote outward or inward remodeling, respectively. However, our understanding of how endothelial cells discern these different flow profiles and regulate gene expression accordingly is limited. This article reviews recent studies that identify the TGFβ/Smad, Notch, Yap/Taz, and Wnt/β-catenin pathways as important mediators of flow profile- and magnitude-dependent signaling.
Collapse
Affiliation(s)
- Elizabeth Min
- Department of Cell Biology, Yale School of Medicine, United States; Yale Cardiovascular Research Center, United States
| | - Martin A Schwartz
- Department of Cell Biology, Yale School of Medicine, United States; Yale Cardiovascular Research Center, United States; Department of Medicine (Cardiology), United States; Department of Biomedical Engineering, United States.
| |
Collapse
|
18
|
Driessen RCH, Stassen OMJA, Sjöqvist M, Suarez Rodriguez F, Grolleman J, Bouten CVC, Sahlgren CM. Shear stress induces expression, intracellular reorganization and enhanced Notch activation potential of Jagged1. Integr Biol (Camb) 2018; 10:719-726. [PMID: 30328449 PMCID: PMC6256362 DOI: 10.1039/c8ib00036k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/15/2018] [Indexed: 01/20/2023]
Abstract
Notch signaling and blood flow regulate vascular formation and maturation, but how shear stress affects the different components of the Notch pathway in endothelial cells is poorly understood. We show that laminar shear stress results in a ligand specific gene expression profile in endothelial cells (HUVEC). JAG1 expression increases while DLL4 expression decreases. Jagged1 shows a unique response by clustering intracellularly six to nine hours after the onset of flow. The formation of the Jagged1 clusters requires protein production, ER export and endocytosis. Clustering is associated with reduced membrane levels but is not affected by Notch signaling activity. Jagged1 relocalization is reversible, the clusters disappear and membrane levels increase upon removal of shear stress. We further demonstrate that the signaling potential of endothelial cells is enhanced after exposure to shear stress. Together we demonstrate a Jagged1 specific shear stress response for Notch signaling in endothelial cells.
Collapse
Affiliation(s)
- R. C. H. Driessen
- Department of Biomedical Engineering, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
.
- Institute for Complex Molecular Systems, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
| | - O. M. J. A. Stassen
- Department of Biomedical Engineering, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
.
| | - M. Sjöqvist
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University
,
Turku
, Finland
| | - F. Suarez Rodriguez
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University
,
Turku
, Finland
| | - J. Grolleman
- Department of Biomedical Engineering, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
.
| | - C. V. C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
.
- Institute for Complex Molecular Systems, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
| | - C. M. Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
.
- Institute for Complex Molecular Systems, Eindhoven University of Technology
,
Eindhoven
, The Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University
,
Turku
, Finland
| |
Collapse
|
19
|
Cerebral ischemia induces angiogenesis in the peri-infarct regions via Notch1 signaling activation. Exp Neurol 2018; 304:30-40. [DOI: 10.1016/j.expneurol.2018.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/13/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
|
20
|
Jones EA, Lehoux S. Shear stress, arterial identity and atherosclerosis. Thromb Haemost 2018; 115:467-73. [DOI: 10.1160/th15-10-0791] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/01/2015] [Indexed: 01/23/2023]
Abstract
SummaryIn the developing embryo, the vasculature first takes the form of a web-like network called the vascular plexus. Arterial and venous differentiation is subsequently guided by the specific expression of genes in the endothelial cells that provide spatial and temporal cues for development. Notch1/4, Notch ligand delta-like 4 (Dll4), and Notch downstream effectors are typically expressed in arterial cells along with EphrinB2, whereas chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) and EphB4 characterise vein endothelial cells. Haemodynamic forces (blood pressure and blood flow) also contribute importantly to vascular remodelling. Early arteriovenous differentiation and local blood flow may hold the key to future inflammatory diseases. Indeed, despite the fact that atherosclerosis risk factors such as smoking, hypertension, hypercholesterolaemia, and diabetes all induce endothelial cell dysfunction throughout the vasculature, plaques develop only in arteries, and they localise essentially in vessel branch points, curvatures and bifurcations, where blood flow (and consequently shear stress) is low or oscillatory. Arterial segments exposed to high blood flow (and high laminar shear stress) tend to remain plaque-free. These observations have led many to investigate what particular properties of arterial or venous endothelial cells confer susceptibility or protection from plaque formation, and how that might interact with a particular shear stress environment.
Collapse
|
21
|
New developments in mechanotransduction: Cross talk of the Wnt, TGF-β and Notch signalling pathways in reaction to shear stress. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2018.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Heuslein JL, McDonnell SP, Song J, Annex BH, Price RJ. MicroRNA-146a Regulates Perfusion Recovery in Response to Arterial Occlusion via Arteriogenesis. Front Bioeng Biotechnol 2018; 6:1. [PMID: 29404323 PMCID: PMC5786509 DOI: 10.3389/fbioe.2018.00001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/03/2018] [Indexed: 01/09/2023] Open
Abstract
The growth of endogenous collateral arteries that bypass arterial occlusion(s), or arteriogenesis, is a fundamental shear stress-induced adaptation with implications for treating peripheral arterial disease. MicroRNAs (miRs) are key regulators of gene expression in response to injury and have strong therapeutic potential. In a previous study, we identified miR-146a as a candidate regulator of vascular remodeling. Here, we tested whether miR-146a regulates in vitro angiogenic endothelial cell (EC) behaviors, as well as perfusion recovery, arteriogenesis, and angiogenesis in response to femoral arterial ligation (FAL) in vivo. We found miR-146a inhibition impaired EC tube formation and migration in vitro. Following FAL, Balb/c mice were treated with a single, intramuscular injection of anti-miR-146a or scramble locked nucleic acid (LNA) oligonucleotides directly into the non-ischemic gracilis muscles. Serial laser Doppler imaging demonstrated that anti-miR-146a treated mice exhibited significantly greater perfusion recovery (a 16% increase) compared mice treated with scramble LNA. Moreover, anti-miR-146a treated mice exhibited a 22% increase in collateral artery diameter compared to controls, while there was no significant effect on in vivo angiogenesis or muscle regeneration. Despite exerting no beneficial effects on angiogenesis, the inhibition of mechanosensitive miR-146a enhances perfusion recovery after FAL via enhanced arteriogenesis.
Collapse
Affiliation(s)
- Joshua L Heuslein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Stephanie P McDonnell
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Ji Song
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Brian H Annex
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
23
|
Sachdev U, Lotze MT. Perpetual change: autophagy, the endothelium, and response to vascular injury. J Leukoc Biol 2017; 102:221-235. [PMID: 28626046 PMCID: PMC6608075 DOI: 10.1189/jlb.3ru1116-484rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Current studies of vascular health, aging, and autophagy emphasize how the endothelium adapts to stress and contributes to disease. The endothelium is far from an inert barrier to blood-borne cells, pathogens, and chemical signals; rather, it actively translates circulating mediators into tissue responses, changing rapidly in response to physiologic stressors. Macroautophagy-the cellular ingestion of effete organelles and protein aggregates to provide anabolic substrates to fuel bioenergetics in times of stress-plays an important role in endothelial cell homeostasis, vascular remodeling, and disease. These roles include regulating vascular tone, sustaining or limiting cell survival, and contributing to the development of atherosclerosis secondary to infection, inflammation, and angiogenesis. Autophagy modulates these critical functions of the endothelium in a dynamic and perpetual response to tissue and intravascular cues.
Collapse
Affiliation(s)
- Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Zhu LP, Zhou JP, Zhang JX, Wang JY, Wang ZY, Pan M, Li LF, Li CC, Wang KK, Bai YP, Zhang GG. MiR-15b-5p Regulates Collateral Artery Formation by Targeting AKT3 (Protein Kinase B-3). Arterioscler Thromb Vasc Biol 2017; 37:957-968. [PMID: 28254819 DOI: 10.1161/atvbaha.116.308905] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 02/16/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To identify circulating microRNAs that are differentially expressed in severe coronary heart disease with well or poorly developed collateral arteries and to investigate their mechanisms of action in vivo and in vitro. APPROACH AND RESULTS In our study, we identified a circulating microRNA, miR-15b-5p, with low expression that, nevertheless, characterized patients with sufficient coronary collateral artery function. Moreover, in murine hindlimb ischemia model, in situ hybridization identified that miR-15b-5p was specifically expressed in vascular endothelial cells of adductors in sham group and was remarkably downregulated after femoral artery ligation. Overexpressed miR-15b-5p significantly inhibited arteriogenesis and angiogenesis in mice. In vitro, both under basal and vascular endothelial growth factor stimulation, loss-of-function or gain-of-function studies suggested that miR-15b-5p significantly promoted or depressed the migration and proliferation of endothelial cells. We identified AKT3 (protein kinase B-3) as a direct target of miR-15b-5p. Interestingly, AKT3 deficiency by injection with Chol-AKT3-siRNA obviously suppressed arteriogenesis and the recovery of blood perfusion after femoral ligation in mice. CONCLUSIONS These results indicate that circulating miR-15b-5p is a suitable biomarker for discriminating between patients with well-developed or poorly developed collaterals. Moreover, miR-15b-5p is a key regulator of arteriogenesis and angiogenesis, which may represent a potential therapeutic target for ischemic disease.
Collapse
Affiliation(s)
- Ling-Ping Zhu
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Ji-Peng Zhou
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Jia-Xiong Zhang
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Jun-Yao Wang
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Zhen-Yu Wang
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Miao Pan
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Ling-Fang Li
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Chuan-Chang Li
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Kang-Kai Wang
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Yong-Ping Bai
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.)
| | - Guo-Gang Zhang
- From the Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (L.-P.Z., J.-P.Z., J.-Y.W., Z.-Y.W., M.P., L.-F.L., L.C., G.-G.Z.); Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China (J.-X.Z., C.-C.L., Y.-P.B.); and Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China (K.-K.W.).
| |
Collapse
|
25
|
Zhou JP, Tong XY, Zhu LP, Luo JM, Luo Y, Bai YP, Li CC, Zhang GG. Plasma Omentin-1 Level as a Predictor of Good Coronary Collateral Circulation. J Atheroscler Thromb 2017; 24:940-948. [PMID: 28123148 PMCID: PMC5587520 DOI: 10.5551/jat.37440] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aims: Coronary collateral circulation (CCC) is crucial during an acute ischemic attack. Evidences showed that omentin-1 exhibited remarkable antiatherogenic effects and ischemia-induced revascularization. The aim of this study was to investigate the relationship between plasma omentin-1 levels and CCC in patients with ≥ 90% angiography-proven coronary occlusion. Methods: 142 patients with ≥ 90% luminal diameter stenosis in at least one major epicardial coronary artery were recruited. Among them, 79 patients with Rentrop 0–1 grade were classified into the poor CCC group and 63 patients with Rentrop 2–3 grade were included into the good CCC group. The association between plasma omentin-1 levels and CCC status was assessed. Results: Plasma omentin-1 level was significantly higher in patients with good CCC than those with poor CCC (566.57 ± 26.90 vs. 492.38 ± 19.70 ng/mL, p = 0.024). Besides, omentin-1 was positively correlated with total cholesterol (TC), high-density lipoprotein, and gensini score but inversely with hyperlipidemia and body mass index (all p values < 0.05). Multivariate regression analysis indicated that omentin-1 [odds ratio (OR) = 1.002, 95% confidence interval (CI): 1.000 – 1.004, p = 0.041)], TC, the number of the diseased vessels, a higher frequency of left circumflex artery and right coronary artery, chronic total occlusion, and gensini score remained as the independent predictors of good CCC. Conclusion: Higher plasma omentin-1 level was associated with better CCC development. Our findings suggest that omentin-1 may be an alternative marker for adequate CCC in patients with ≥ 90% coronary occlusion.
Collapse
Affiliation(s)
- Ji-Peng Zhou
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University
| | - Xiao-Yu Tong
- Department of Geriatric Medicine, Xiangya Hospital, Central South University
| | - Ling-Ping Zhu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University
| | - Jing-Min Luo
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University
| | - Ying Luo
- Department of Geriatric Medicine, Xiangya Hospital, Central South University
| | - Yong-Ping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University
| | - Chuan-Chang Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University
| | - Guo-Gang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University
| |
Collapse
|
26
|
Abstract
SIGNIFICANCE Forces are important in the cardiovascular system, acting as regulators of vascular physiology and pathology. Residing at the blood vessel interface, cells (endothelial cell, EC) are constantly exposed to vascular forces, including shear stress. Shear stress is the frictional force exerted by blood flow, and its patterns differ based on vessel geometry and type. These patterns range from uniform laminar flow to nonuniform disturbed flow. Although ECs sense and differentially respond to flow patterns unique to their microenvironment, the mechanisms underlying endothelial mechanosensing remain incompletely understood. RECENT ADVANCES A large body of work suggests that ECs possess many mechanosensors that decorate their apical, junctional, and basal surfaces. These potential mechanosensors sense blood flow, translating physical force into biochemical signaling events. CRITICAL ISSUES Understanding the mechanisms by which proposed mechanosensors sense and respond to shear stress requires an integrative approach. It is also critical to understand the role of these mechanosensors not only during embryonic development but also in the different vascular beds in the adult. Possible cross talk and integration of mechanosensing via the various mechanosensors remain a challenge. FUTURE DIRECTIONS Determination of the hierarchy of endothelial mechanosensors is critical for future work, as is determination of the extent to which mechanosensors work together to achieve force-dependent signaling. The role and primary sensors of shear stress during development also remain an open question. Finally, integrative approaches must be used to determine absolute mechanosensory function of potential mechanosensors. Antioxid. Redox Signal. 25, 373-388.
Collapse
Affiliation(s)
- Chris Givens
- 1 Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina
| | - Ellie Tzima
- 1 Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina.,2 Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics , Oxford, United Kingdom
| |
Collapse
|
27
|
Yurdagul A, Orr AW. Blood Brothers: Hemodynamics and Cell-Matrix Interactions in Endothelial Function. Antioxid Redox Signal 2016; 25:415-34. [PMID: 26715135 PMCID: PMC5011636 DOI: 10.1089/ars.2015.6525] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/25/2015] [Accepted: 12/23/2015] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Alterations in endothelial function contribute to a variety of vascular diseases. In pathological conditions, the endothelium shows a reduced ability to regulate vasodilation (endothelial dysfunction) and a conversion toward a proinflammatory and leaky phenotype (endothelial activation). At the interface between the vessel wall and blood, the endothelium exists in a complex microenvironment and must translate changes in these environmental signals to alterations in vessel function. Mechanical stimulation and endothelial cell interactions with the vascular matrix, as well as a host of soluble factors, coordinately contribute to this dynamic regulation. RECENT ADVANCES Blood hemodynamics play an established role in the regulation of endothelial function. However, a growing body of work suggests that subendothelial matrix composition similarly and coordinately regulates endothelial cell phenotype such that blood flow affects matrix remodeling, which affects the endothelial response to flow. CRITICAL ISSUES Hemodynamics and soluble factors likely affect endothelial matrix remodeling through multiple mechanisms, including transforming growth factor β signaling and alterations in cell-matrix receptors, such as the integrins. Likewise, differential integrin signaling following matrix remodeling appears to regulate several key flow-induced responses, including nitric oxide production, regulation of oxidant stress, and activation of proinflammatory signaling and gene expression. Microvascular remodeling responses, such as angiogenesis and arteriogenesis, may also show coordinated regulation by flow and matrix. FUTURE DIRECTIONS Identifying the mechanisms regulating the dynamic interplay between hemodynamics and matrix remodeling and their contribution to the pathogenesis of cardiovascular disease remains an important research area with therapeutic implications across a variety of conditions. Antioxid. Redox Signal. 25, 415-434.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| | - A. Wayne Orr
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| |
Collapse
|
28
|
Zhu H, Zhang M, Liu Z, Xing J, Moriasi C, Dai X, Zou MH. AMP-Activated Protein Kinase α1 in Macrophages Promotes Collateral Remodeling and Arteriogenesis in Mice In Vivo. Arterioscler Thromb Vasc Biol 2016; 36:1868-78. [PMID: 27444205 DOI: 10.1161/atvbaha.116.307743] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/27/2016] [Indexed: 01/12/2023]
Abstract
OBJECTIVE AMP-activated protein kinase (AMPK), an energy and redox sensor, is activated in response to various cellular stresses, including hypoxia, nutrient deprivation, oxidative stress, and fluid shear stress at the site of vessel blockade. The activation of AMPK is involved in angiogenesis. However, it is unknown whether AMPK can influence arteriogenesis. Here, we demonstrate the contribution of macrophage AMPK to arteriogenesis and collateral remodeling and their underlying mechanisms in well-characterized in vivo and in vitro models. APPROACH AND RESULTS AMPKα1, AMPKα2 knockout and wild-type littermates underwent femoral artery ligation. Collateral arteriogenesis was monitored in wild-type, global AMPKα1 knockout, or macrophage-specific AMPKα1 knockout mice, with or without hindlimb ligation. Compared with wild-type mice with ligation, global AMPKα1 knockout mice displayed significant reduction in blood flow recovery and impaired remodeling of collateral arterioles. Similar impairments were observed in macrophage-specific AMPK α1 knockout mice after hindlimb ligation. Mechanistically, we found that AMPKα1 promotes the production of growth factors, such as transforming growth factor β, by directly phosphorylating the inhibitor of nuclear factor κB kinase alpha, resulting in an nuclear factor κB-dependent production of growth factors CONCLUSIONS Our findings suggest a novel role for macrophage AMPKα1 in arteriogenesis and collateral remodeling and indicate that AMPKα1 activation might be beneficial for recovery from occlusive vascular disorders.
Collapse
Affiliation(s)
- Huaiping Zhu
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.)
| | - Miao Zhang
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.)
| | - Zhaoyu Liu
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.)
| | - Junjie Xing
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.)
| | - Cate Moriasi
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.)
| | - Xiaoyan Dai
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.)
| | - Ming-Hui Zou
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (H.Z., Z.L., C.M., X.D., M.-H.Z.); and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z., J.X.).
| |
Collapse
|
29
|
Samsa LA, Givens C, Tzima E, Stainier DYR, Qian L, Liu J. Cardiac contraction activates endocardial Notch signaling to modulate chamber maturation in zebrafish. Development 2016; 142:4080-91. [PMID: 26628092 DOI: 10.1242/dev.125724] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Congenital heart disease often features structural abnormalities that emerge during development. Accumulating evidence indicates a crucial role for cardiac contraction and the resulting fluid forces in shaping the heart, yet the molecular basis of this function is largely unknown. Using the zebrafish as a model of early heart development, we investigated the role of cardiac contraction in chamber maturation, focusing on the formation of muscular protrusions called trabeculae. By genetic and pharmacological ablation of cardiac contraction, we showed that cardiac contraction is required for trabeculation through its role in regulating notch1b transcription in the ventricular endocardium. We also showed that Notch1 activation induces expression of ephrin b2a (efnb2a) and neuregulin 1 (nrg1) in the endocardium to promote trabeculation and that forced Notch activation in the absence of cardiac contraction rescues efnb2a and nrg1 expression. Using in vitro and in vivo systems, we showed that primary cilia are important mediators of fluid flow to stimulate Notch expression. Together, our findings describe an essential role for cardiac contraction-responsive transcriptional changes in endocardial cells to regulate cardiac chamber maturation.
Collapse
Affiliation(s)
- Leigh Ann Samsa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chris Givens
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eleni Tzima
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Li Qian
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
Heuslein JL, Meisner JK, Li X, Song J, Vincentelli H, Leiphart RJ, Ames EG, Blackman BR, Blackman BR, Price RJ. Mechanisms of Amplified Arteriogenesis in Collateral Artery Segments Exposed to Reversed Flow Direction. Arterioscler Thromb Vasc Biol 2015; 35:2354-65. [PMID: 26338297 PMCID: PMC4618717 DOI: 10.1161/atvbaha.115.305775] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/14/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Collateral arteriogenesis, the growth of existing arterial vessels to a larger diameter, is a fundamental adaptive response that is often critical for the perfusion and survival of tissues downstream of chronic arterial occlusion(s). Shear stress regulates arteriogenesis; however, the arteriogenic significance of reversed flow direction, occurring in numerous collateral artery segments after femoral artery ligation, is unknown. Our objective was to determine if reversed flow direction in collateral artery segments differentially regulates endothelial cell signaling and arteriogenesis. APPROACH AND RESULTS Collateral segments experiencing reversed flow direction after femoral artery ligation in C57BL/6 mice exhibit increased pericollateral macrophage recruitment, amplified arteriogenesis (30% diameter and 2.8-fold conductance increases), and remarkably permanent (12 weeks post femoral artery ligation) remodeling. Genome-wide transcriptional analyses on human umbilical vein endothelial cells exposed to reversed flow conditions mimicking those occurring in vivo yielded 10-fold more significantly regulated transcripts, as well as enhanced activation of upstream regulators (nuclear factor κB [NFκB], vascular endothelial growth factor, fibroblast growth factor-2, and transforming growth factor-β) and arteriogenic canonical pathways (protein kinase A, phosphodiesterase, and mitogen-activated protein kinase). Augmented expression of key proarteriogenic molecules (Kruppel-like factor 2 [KLF2], intercellular adhesion molecule 1, and endothelial nitric oxide synthase) was also verified by quantitative real-time polymerase chain reaction, leading us to test whether intercellular adhesion molecule 1 or endothelial nitric oxide synthase regulate amplified arteriogenesis in flow-reversed collateral segments in vivo. Interestingly, enhanced pericollateral macrophage recruitment and amplified arteriogenesis was attenuated in flow-reversed collateral segments after femoral artery ligation in intercellular adhesion molecule 1(-/-) mice; however, endothelial nitric oxide synthase(-/-) mice showed no such differences. CONCLUSIONS Reversed flow leads to a broad amplification of proarteriogenic endothelial signaling and a sustained intercellular adhesion molecule 1-dependent augmentation of arteriogenesis. Further investigation of the endothelial mechanotransduction pathways activated by reversed flow may lead to more effective and durable therapeutic options for arterial occlusive diseases.
Collapse
Affiliation(s)
- Joshua L Heuslein
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Joshua K Meisner
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Xuanyue Li
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Ji Song
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Helena Vincentelli
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Ryan J Leiphart
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Elizabeth G Ames
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Brett R Blackman
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | | | - Richard J Price
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.).
| |
Collapse
|
31
|
Vandersmissen I, Craps S, Depypere M, Coppiello G, van Gastel N, Maes F, Carmeliet G, Schrooten J, Jones EAV, Umans L, Devlieger R, Koole M, Gheysens O, Zwijsen A, Aranguren XL, Luttun A. Endothelial Msx1 transduces hemodynamic changes into an arteriogenic remodeling response. J Cell Biol 2015; 210:1239-56. [PMID: 26391659 PMCID: PMC4586738 DOI: 10.1083/jcb.201502003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 08/18/2015] [Indexed: 12/25/2022] Open
Abstract
During peripheral arterial disease, MSX1 acts downstream of BMP–SMAD signaling to transduce the arterial shear stimulus into an arteriogenic remodeling response. MSX1 activates collateral endothelium into a proinflammatory state through ICAM1/VCAM1 up-regulation, resulting in increased leukocyte infiltration and collateral remodeling. Collateral remodeling is critical for blood flow restoration in peripheral arterial disease and is triggered by increasing fluid shear stress in preexisting collateral arteries. So far, no arterial-specific mediators of this mechanotransduction response have been identified. We show that muscle segment homeobox 1 (MSX1) acts exclusively in collateral arterial endothelium to transduce the extrinsic shear stimulus into an arteriogenic remodeling response. MSX1 was specifically up-regulated in remodeling collateral arteries. MSX1 induction in collateral endothelial cells (ECs) was shear stress driven and downstream of canonical bone morphogenetic protein–SMAD signaling. Flow recovery and collateral remodeling were significantly blunted in EC-specific Msx1/2 knockout mice. Mechanistically, MSX1 linked the arterial shear stimulus to arteriogenic remodeling by activating the endothelial but not medial layer to a proinflammatory state because EC but not smooth muscle cellMsx1/2 knockout mice had reduced leukocyte recruitment to remodeling collateral arteries. This reduced leukocyte infiltration in EC Msx1/2 knockout mice originated from decreased levels of intercellular adhesion molecule 1 (ICAM1)/vascular cell adhesion molecule 1 (VCAM1), whose expression was also in vitro driven by promoter binding of MSX1.
Collapse
Affiliation(s)
- Ine Vandersmissen
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Sander Craps
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Maarten Depypere
- Department of Electrical Engineering/Processing Speech and Images, Medical Image Computing, KU Leuven, 3000 Leuven, Belgium
| | - Giulia Coppiello
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Division of Skeletal Tissue Engineering, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Frederik Maes
- Department of Electrical Engineering/Processing Speech and Images, Medical Image Computing, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Division of Skeletal Tissue Engineering, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Jan Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium Department of Materials Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Umans
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Roland Devlieger
- Department of Gynecology and Obstetrics, University Hospital Leuven, 3000 Leuven, Belgium Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Olivier Gheysens
- Department of Nuclear Medicine University Hospital Leuven, 3000 Leuven, Belgium Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - An Zwijsen
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Xabier L Aranguren
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
| | - Aernout Luttun
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
32
|
Paik DT, Rai M, Ryzhov S, Sanders LN, Aisagbonhi O, Funke MJ, Feoktistov I, Hatzopoulos AK. Wnt10b Gain-of-Function Improves Cardiac Repair by Arteriole Formation and Attenuation of Fibrosis. Circ Res 2015; 117:804-16. [PMID: 26338900 DOI: 10.1161/circresaha.115.306886] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/03/2015] [Indexed: 01/10/2023]
Abstract
RATIONALE Myocardial infarction causes irreversible tissue damage, leading to heart failure. We recently discovered that canonical Wnt signaling and the Wnt10b ligand are strongly induced in mouse hearts after infarction. Wnt10b regulates cell fate in various organs, but its role in the heart is unknown. OBJECTIVE To investigate the effect of Wnt10b gain-of-function on cardiac repair mechanisms and to assess its potential to improve ventricular function after injury. METHODS AND RESULTS Histological and molecular analyses showed that Wnt10b is expressed in cardiomyocytes and localized in the intercalated discs of mouse and human hearts. After coronary artery ligation or cryoinjury in mice, Wnt10b is strongly and transiently induced in peri-infarct cardiomyocytes during granulation tissue formation. To determine the effect of Wnt10b on neovascularization and fibrosis, we generated a mouse line to increase endogenous Wnt10b levels in cardiomyocytes. We found that gain of Wnt10b function orchestrated a recovery phenotype characterized by robust neovascularization of the injury zone, less myofibroblasts, reduced scar size, and improved ventricular function compared with wild-type mice. Wnt10b stimulated expression of vascular endothelial growth factor receptor 2 in endothelial cells and angiopoietin-1 in vascular smooth muscle cells through nuclear factor-κB activation. These effects coordinated endothelial growth and smooth muscle cell recruitment, promoting robust formation of large, coronary-like blood vessels. CONCLUSION Wnt10b gain-of-function coordinates arterial formation and attenuates fibrosis in cardiac tissue after injury. Because generation of mature blood vessels is necessary for efficient perfusion, our findings could lead to novel strategies to optimize the inherent repair capacity of the heart and prevent the onset of heart failure.
Collapse
Affiliation(s)
- David T Paik
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Meena Rai
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Sergey Ryzhov
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Lehanna N Sanders
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Omonigho Aisagbonhi
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Mitchell J Funke
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Igor Feoktistov
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Antonis K Hatzopoulos
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.).
| |
Collapse
|
33
|
Sweet DT, Jiménez JM, Chang J, Hess PR, Mericko-Ishizuka P, Fu J, Xia L, Davies PF, Kahn ML. Lymph flow regulates collecting lymphatic vessel maturation in vivo. J Clin Invest 2015. [PMID: 26214523 DOI: 10.1172/jci79386] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Fluid shear forces have established roles in blood vascular development and function, but whether such forces similarly influence the low-flow lymphatic system is unknown. It has been difficult to test the contribution of fluid forces in vivo because mechanical or genetic perturbations that alter flow often have direct effects on vessel growth. Here, we investigated the functional role of flow in lymphatic vessel development using mice deficient for the platelet-specific receptor C-type lectin-like receptor 2 (CLEC2) as blood backfills the lymphatic network and blocks lymph flow in these animals. CLEC2-deficient animals exhibited normal growth of the primary mesenteric lymphatic plexus but failed to form valves in these vessels or remodel them into a structured, hierarchical network. Smooth muscle cell coverage (SMC coverage) of CLEC2-deficient lymphatic vessels was both premature and excessive, a phenotype identical to that observed with loss of the lymphatic endothelial transcription factor FOXC2. In vitro evaluation of lymphatic endothelial cells (LECs) revealed that low, reversing shear stress is sufficient to induce expression of genes required for lymphatic valve development and identified GATA2 as an upstream transcriptional regulator of FOXC2 and the lymphatic valve genetic program. These studies reveal that lymph flow initiates and regulates many of the key steps in collecting lymphatic vessel maturation and development.
Collapse
|
34
|
Baeyens N, Nicoli S, Coon BG, Ross TD, Van den Dries K, Han J, Lauridsen HM, Mejean CO, Eichmann A, Thomas JL, Humphrey JD, Schwartz MA. Vascular remodeling is governed by a VEGFR3-dependent fluid shear stress set point. eLife 2015; 4. [PMID: 25643397 PMCID: PMC4337723 DOI: 10.7554/elife.04645] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 02/01/2015] [Indexed: 12/23/2022] Open
Abstract
Vascular remodeling under conditions of growth or exercise, or during recovery from arterial restriction or blockage is essential for health, but mechanisms are poorly understood. It has been proposed that endothelial cells have a preferred level of fluid shear stress, or ‘set point’, that determines remodeling. We show that human umbilical vein endothelial cells respond optimally within a range of fluid shear stress that approximate physiological shear. Lymphatic endothelial cells, which experience much lower flow in vivo, show similar effects but at lower value of shear stress. VEGFR3 levels, a component of a junctional mechanosensory complex, mediate these differences. Experiments in mice and zebrafish demonstrate that changing levels of VEGFR3/Flt4 modulates aortic lumen diameter consistent with flow-dependent remodeling. These data provide direct evidence for a fluid shear stress set point, identify a mechanism for varying the set point, and demonstrate its relevance to vessel remodeling in vivo. DOI:http://dx.doi.org/10.7554/eLife.04645.001 Blood and lymphatic vessels remodel their shape, diameter and connections during development, and throughout life in response to growth, exercise and disease. This process is called vascular remodeling. The endothelial cells that line the inside of blood and lymphatic vessels are constantly exposed to the frictional force from flowing blood, termed fluid shear stress. Changes in shear stress are sensed by the endothelial cells, which trigger vascular remodeling to return the stress to the original level. It has been proposed that remodeling is governed by a preferred level of fluid shear stress, or set point, against which deviations in the shear stress are compared. Thus, changing the fluid flow through a blood vessel increases or decreases shear stress, which results in the vessel remodeling to restore the original level of shear stress. Like all remodeling, this process involves inflammation to recruit white blood cells, which assist with the process. Baeyens et al. investigated whether such a shear stress set point exists and what its biological basis might be using cultured endothelial cells from human umbilical veins. These cells remained stable and in a resting state when a particular level of shear stress was applied to them; above or below this shear stress level, the cells produced an inflammatory response like that seen during vascular remodeling. This suggests that these cells do indeed have a set point for shear stress. The same response occurred in human lymphatic endothelial cells, although in these cells the shear stress set point was much lower, correlating with the low flow in lymphatic vessels. Baeyens et al. then discovered that the shear stress set point is related to the level of a protein called VEGFR3 in the cells, which was recently found to participate in shear stress sensing. Endothelial cells from lymphatic vessels normally produce much greater quantities of VEGFR3 than those from blood vessels. Reducing the amount of VEGFR3 in lymphatic endothelial cells increased the set point shear stress, while increasing the levels in blood vessel cells decreased the set point. This suggests that the levels of this protein account for the difference in the response of these two cell types. Baeyens et al. then tested this pathway by reducing the levels of VEGFR3 in zebrafish embryos and in adult mice. In both animals, this caused arteries to narrow, showing that VEGFR3 levels also control sensitivity to shear stress—and hence vascular remodeling—inside living creatures. Understanding in detail how vascular remodeling is regulated could help improve treatments for a wide range of cardiovascular conditions. To do so, further work will be needed to develop methods to control the sensitivity of endothelial cells to shear stress and to identify other proteins that might specifically control the narrowing or the expansion of vessels in human patients. DOI:http://dx.doi.org/10.7554/eLife.04645.002
Collapse
Affiliation(s)
- Nicolas Baeyens
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Stefania Nicoli
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Brian G Coon
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Tyler D Ross
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Koen Van den Dries
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Jinah Han
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Holly M Lauridsen
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, United States
| | - Cecile O Mejean
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Anne Eichmann
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, United States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, United States
| | - Martin A Schwartz
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
35
|
Rizzo P, Mele D, Caliceti C, Pannella M, Fortini C, Clementz AG, Morelli MB, Aquila G, Ameri P, Ferrari R. The role of notch in the cardiovascular system: potential adverse effects of investigational notch inhibitors. Front Oncol 2015; 4:384. [PMID: 25629006 PMCID: PMC4292456 DOI: 10.3389/fonc.2014.00384] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022] Open
Abstract
Targeting the Notch pathway is a new promising therapeutic approach for cancer patients. Inhibition of Notch is effective in the oncology setting because it causes a reduction of highly proliferative tumor cells and it inhibits survival of cancer stem cells, which are considered responsible for tumor recurrence and metastasis. Additionally, since Delta-like ligand 4 (Dll4)-activated Notch signaling is a major modulator of angiogenesis, anti-Dll4 agents are being investigated to reduce vascularization of the tumor. Notch plays a major role in the heart during the development and, after birth, in response to cardiac damage. Therefore, agents used to inhibit Notch in the tumors (gamma secretase inhibitors and anti-Dll4 agents) could potentially affect myocardial repair. The past experience with trastuzumab and other tyrosine kinase inhibitors used for cancer therapy demonstrates that the possible cardiotoxicity of agents targeting shared pathways between cancer and heart and the vasculature should be considered. To date, Notch inhibition in cancer patients has resulted only in mild gastrointestinal toxicity. Little is known about the potential long-term cardiotoxicity associated to Notch inhibition in cancer patients. In this review, we will focus on mechanisms through which inhibition of Notch signaling could lead to cardiomyocytes and endothelial dysfunctions. These adverse effects could contrast with the benefits of therapeutic responses in cancer cells during times of increased cardiac stress and/or in the presence of cardiovascular risk factor.
Collapse
Affiliation(s)
- Paola Rizzo
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy ; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy ; GVM Hospitals , Cotignola , Italy
| | - Donato Mele
- Azienda Ospedaliero-Universitaria di Ferrara , Cona , Italy
| | | | - Micaela Pannella
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Cinzia Fortini
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | | | | | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Pietro Ameri
- Research Center of Cardiovascular Biology, Department of Internal Medicine, University of Genova , Genova , Italy
| | - Roberto Ferrari
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy ; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy ; Azienda Ospedaliero-Universitaria di Ferrara , Cona , Italy
| |
Collapse
|
36
|
|