1
|
Deng H, Rukhlenko OS, Joshi D, Hu X, Junk P, Tuliakova A, Kholodenko BN, Schwartz MA. cSTAR analysis identifies endothelial cell cycle as a key regulator of flow-dependent artery remodeling. SCIENCE ADVANCES 2025; 11:eado9970. [PMID: 39752487 PMCID: PMC11698091 DOI: 10.1126/sciadv.ado9970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025]
Abstract
Fluid shear stress (FSS) from blood flow sensed by vascular endothelial cells (ECs) determines vessel behavior, but regulatory mechanisms are only partially understood. We used cell state transition assessment and regulation (cSTAR), a powerful computational method, to elucidate EC transcriptomic states under low shear stress (LSS), physiological shear stress (PSS), high shear stress (HSS), and oscillatory shear stress (OSS) that induce vessel inward remodeling, stabilization, outward remodeling, or disease susceptibility, respectively. Combined with a publicly available database on EC transcriptomic responses to drug treatments, this approach inferred a regulatory network controlling EC states and made several notable predictions. Particularly, inhibiting cell cycle-dependent kinase (CDK) 2 was predicted to initiate inward remodeling and promote atherogenesis. In vitro, PSS activated CDK2 and induced late G1 cell cycle arrest. In mice, EC deletion of CDK2 triggered inward artery remodeling, pulmonary and systemic hypertension, and accelerated atherosclerosis. These results validate use of cSTAR and identify key determinants of normal and pathological artery remodeling.
Collapse
Affiliation(s)
- Hanqiang Deng
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
| | - Oleksii S. Rukhlenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Divyesh Joshi
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA
| | - Xiaoyue Hu
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
| | - Philipp Junk
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Anna Tuliakova
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Boris N. Kholodenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT 06510, USA
| |
Collapse
|
2
|
Haritz JL, Pflaum M, Güntner HJ, Katsirntaki K, Hegermann J, Hehnen F, Lommel M, Kertzscher U, Arens J, Haverich A, Ruhparwar A, Wiegmann B. Citrate-Coated Iron Oxide Nanoparticles Facilitate Endothelialization of Left Ventricular Assist Device Impeller for Improved Antithrombogenicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408976. [PMID: 39707689 DOI: 10.1002/advs.202408976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/14/2024] [Indexed: 12/23/2024]
Abstract
Although left ventricular assist devices (LVADs) are an alternative to heart transplantation, their artificial surfaces often lead to serious thrombotic complications requiring high-risk device replacement. Coating blood-contacting surfaces with antithrombogenic endothelial cells is considered an effective strategy for preventing thrombus formation. However, this concept has not yet been successfully implemented in LVADs, as severe cell loss is to be expected, especially on the impeller surface with high prothrombogenic supraphysiological shear stress. This study presents a strategy that exploits the magnetic attraction of the impeller on ECs loaded with iron oxide nanoparticles (IONPs) to minimize shear stress-induced cell detachment from the rotating magnetic impeller while ensuring antithrombogenic EC adhesion, especially as a bridge until they formed their adhesion-promoting matrix. In contrast to polyvinylpyrrolidone (PVP)-coated IONPs, more efficient and safer cell loading is achieved with sodium citrate (Cit)-stabilized IONPs, where incubation with 6.6 µg iron mL-1 Cit-IONPs for 24 h resulting in an average internalization of 23 pg iron per cell. Internalization of Cit-IONP significantly improved cell attraction to the highly magnetic impeller surface without affecting cell viability or antithrombogenic function. This protocol is key for the development of a biohybrid LVAD impeller that can prevent life-threatening thrombosis and hemorrhage in a future clinical application.
Collapse
Affiliation(s)
- Jasper L Haritz
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
| | - Michael Pflaum
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans J Güntner
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
| | - Katherina Katsirntaki
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
| | - Jan Hegermann
- German Center for Lung Research, BREATH, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Research Core Unit Electron Microscopy and Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Felix Hehnen
- Biofluid Mechanics Laboratory, Institute of Computer-assisted Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Michael Lommel
- Biofluid Mechanics Laboratory, Institute of Computer-assisted Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Ulrich Kertzscher
- Biofluid Mechanics Laboratory, Institute of Computer-assisted Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jutta Arens
- Engineering Organ Support Technologies Group, Department of Biomechanical Engineering, Faculty of Engineering Technology, University of Twente, Enschede, NB, 7522, Netherlands
- Member of the DFG-SPP2014, 30625, Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Arjang Ruhparwar
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Bettina Wiegmann
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Member of the DFG-SPP2014, 30625, Hannover, Germany
| |
Collapse
|
3
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
4
|
Wang Y, Liu M, Zhang W, Liu H, Jin F, Mao S, Han C, Wang X. Mechanical strategies to promote vascularization for tissue engineering and regenerative medicine. BURNS & TRAUMA 2024; 12:tkae039. [PMID: 39350780 PMCID: PMC11441985 DOI: 10.1093/burnst/tkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 10/04/2024]
Abstract
Vascularization is a major challenge in the field of tissue engineering and regenerative medicine. Mechanical factors have been demonstrated to play a fundamental role in vasculogenesis and angiogenesis and can affect the architecture of the generated vascular network. Through the regulation of mechanical factors in engineered tissues, various mechanical strategies can be used to optimize the preformed vascular network and promote its rapid integration with host vessels. Optimization of the mechanical properties of scaffolds, including controlling scaffold stiffness, increasing surface roughness and anisotropic structure, and designing interconnected, hierarchical pore structures, is beneficial for the in vitro formation of vascular networks and the ingrowth of host blood vessels. The incorporation of hollow channels into scaffolds promotes the formation of patterned vascular networks. Dynamic stretching and perfusion can facilitate the formation and maturation of preformed vascular networks in vitro. Several indirect mechanical strategies provide sustained mechanical stimulation to engineered tissues in vivo, which further promotes the vascularization of implants within the body. Additionally, stiffness gradients, anisotropic substrates and hollow channels in scaffolds, as well as external cyclic stretch, boundary constraints and dynamic flow culture, can effectively regulate the alignment of vascular networks, thereby promoting better integration of prevascularized engineered tissues with host blood vessels. This review summarizes the influence and contribution of both scaffold-based and external stimulus-based mechanical strategies for vascularization in tissue engineering and elucidates the underlying mechanisms involved.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Meixuan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Huan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Fang Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Shulei Mao
- Department of Burns and Plastic Surgery, Quhua Hospital of Zhejiang, 62 Wenchang Road, Quhua, Quzhou 324004, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| |
Collapse
|
5
|
Zisser L, Binder CJ. Extracellular Vesicles as Mediators in Atherosclerotic Cardiovascular Disease. J Lipid Atheroscler 2024; 13:232-261. [PMID: 39355407 PMCID: PMC11439751 DOI: 10.12997/jla.2024.13.3.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/03/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial intima, characterized by accumulation of lipoproteins and accompanying inflammation, leading to the formation of plaques that eventually trigger occlusive thrombotic events, such as myocardial infarction and ischemic stroke. Although many aspects of plaque development have been elucidated, the role of extracellular vesicles (EVs), which are lipid bilayer-delimited vesicles released by cells as mediators of intercellular communication, has only recently come into focus of atherosclerosis research. EVs comprise several subtypes that may be differentiated by their size, mode of biogenesis, or surface marker expression and cargo. The functional effects of EVs in atherosclerosis depend on their cellular origin and the specific pathophysiological context. EVs have been suggested to play a role in all stages of plaque formation. In this review, we highlight the known mechanisms by which EVs modulate atherogenesis and outline current limitations and challenges in the field.
Collapse
Affiliation(s)
- Lucia Zisser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Wang LH, Yang B, Wang Z, Jia L, Chen HY, Bi XQ. Effects of tanshinone IIA on endothelial cell dysfunction in uremic condition. J Biochem Mol Toxicol 2024; 38:e23785. [PMID: 39051181 DOI: 10.1002/jbt.23785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/27/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
An arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis in uremic patients, yet its dysfunction poses a significant clinical challenge. Venous stenosis, primarily caused by venous neointimal hyperplasia, is a key factor in the failure of vascular access. During vascular access dysfunction, endothelial cells (ECs) transform mechanical stimuli into intracellular signals and interact with vascular smooth muscle cells. Tanshinone IIA, an important compound derived from Salvia miltiorrhiza, has been widely used to treat cardiovascular diseases. However, its role in modulating ECs under uremic conditions remains incompletely understood. In this research, ECs were exposed to sodium tanshinone IIA sulfonate (STS) and subjected to shear stress and uremic conditions. The results indicate that STS can reduce the suppressive effects on the expression of NF-κB p65, JNK and Collagen I in uremia-induced ECs. Moreover, the downregulation of NF-κB p65, JNK and Collagen I can be enhanced through the inhibition of ERK1/2 and the upregulation of Caveolin-1. These findings suggest that tanshinone IIA may improve EC function under uremic conditions by targeting the Caveolin-1/ERK1/2 pathway, presenting tanshinone IIA as a potential therapeutic agent against AVF immaturity caused by EC dysfunction.
Collapse
Affiliation(s)
- Li-Hua Wang
- Department of Kidney Disease and Blood Purification Center, 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Bo Yang
- Department of Nephrology, The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhe Wang
- Department of Kidney Disease and Blood Purification Center, 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Lan Jia
- Department of Kidney Disease and Blood Purification Center, 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Hai-Yan Chen
- Department of Kidney Disease and Blood Purification Center, 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Xue-Qing Bi
- Department of Kidney Disease and Blood Purification Center, 2nd Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
7
|
Min L, Zhong F, Gu L, Lee K, He JC. Krüppel-like factor 2 is an endoprotective transcription factor in diabetic kidney disease. Am J Physiol Cell Physiol 2024; 327:C477-C486. [PMID: 38981608 DOI: 10.1152/ajpcell.00222.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Diabetic kidney disease (DKD) is a microvascular complication of diabetes, and glomerular endothelial cell (GEC) dysfunction is a key driver of DKD pathogenesis. Krüppel-like factor 2 (KLF2), a shear stress-induced transcription factor, is among the highly regulated genes in early DKD. In the kidney, KLF2 expression is mostly restricted to endothelial cells, but its expression is also found in immune cell subsets. KLF2 expression is upregulated in response to increased shear stress by the activation of mechanosensory receptors but suppressed by inflammatory cytokines, both of which characterize the early diabetic kidney milieu. KLF2 expression is reduced in progressive DKD and hypertensive nephropathy in humans and mice, likely due to high glucose and inflammatory cytokines such as TNF-α. However, KLF2 expression is increased in glomerular hyperfiltration-induced shear stress without metabolic dysregulation, such as in settings of unilateral nephrectomy. Lower KLF2 expression is associated with CKD progression in patients with unilateral nephrectomy, consistent with its endoprotective role. KLF2 confers endoprotection by inhibition of inflammation, thrombotic activation, and angiogenesis, and thus KLF2 is considered a protective factor for cardiovascular disease (CVD). Based on similar mechanisms, KLF2 also exhibits renoprotection, and its reduced expression in endothelial cells worsens glomerular injury and albuminuria in settings of diabetes or unilateral nephrectomy. Thus KLF2 confers endoprotective effects in both CVD and DKD, and its activators could potentially be developed as a novel class of drugs for cardiorenal protection in diabetic patients.
Collapse
Affiliation(s)
- Lulin Min
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Fang Zhong
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kyung Lee
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - John Cijiang He
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Renal Section, James J. Peters Veterans Affair Medical Center, Bronx, New York, United States
| |
Collapse
|
8
|
Ning L, Zanella S, Tomov ML, Amoli MS, Jin L, Hwang B, Saadeh M, Chen H, Neelakantan S, Dasi LP, Avazmohammadi R, Mahmoudi M, Bauser‐Heaton HD, Serpooshan V. Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400476. [PMID: 38696618 PMCID: PMC11234432 DOI: 10.1002/advs.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/09/2024] [Indexed: 05/04/2024]
Abstract
Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.
Collapse
Affiliation(s)
- Liqun Ning
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of Mechanical EngineeringCleveland State UniversityClevelandOH44115USA
| | - Stefano Zanella
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Martin L. Tomov
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Linqi Jin
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Boeun Hwang
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Maher Saadeh
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Huang Chen
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Sunder Neelakantan
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lakshmi Prasad Dasi
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Reza Avazmohammadi
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
- J. Mike Walker ’66 Department of Mechanical EngineeringTexas A&M UniversityCollege StationTX77840USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LandingMI48824USA
| | - Holly D. Bauser‐Heaton
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
- Sibley Heart Center at Children's Healthcare of AtlantaAtlantaGA30322USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
| |
Collapse
|
9
|
Berg K, Gorham J, Lundt F, Seidman J, Brueckner M. Endocardial primary cilia and blood flow are required for regulation of EndoMT during endocardial cushion development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594405. [PMID: 38798559 PMCID: PMC11118576 DOI: 10.1101/2024.05.15.594405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Blood flow is critical for heart valve formation, and cellular mechanosensors are essential to translate flow into transcriptional regulation of development. Here, we identify a role for primary cilia in vivo in the spatial regulation of cushion formation, the first stage of valve development, by regionally controlling endothelial to mesenchymal transition (EndoMT) via modulation of Kruppel-like Factor 4 (Klf4) . We find that high shear stress intracardiac regions decrease endocardial ciliation over cushion development, correlating with KLF4 downregulation and EndoMT progression. Mouse embryos constitutively lacking cilia exhibit a blood-flow dependent accumulation of KLF4 in these regions, independent of upstream left-right abnormalities, resulting in impaired cushion cellularization. snRNA-seq revealed that cilia KO endocardium fails to progress to late-EndoMT, retains endothelial markers and has reduced EndoMT/mesenchymal genes that KLF4 antagonizes. Together, these data identify a mechanosensory role for endocardial primary cilia in cushion development through regional regulation of KLF4.
Collapse
|
10
|
Abdelilah-Seyfried S, Ola R. Shear stress and pathophysiological PI3K involvement in vascular malformations. J Clin Invest 2024; 134:e172843. [PMID: 38747293 PMCID: PMC11093608 DOI: 10.1172/jci172843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
Molecular characterization of vascular anomalies has revealed that affected endothelial cells (ECs) harbor gain-of-function (GOF) mutations in the gene encoding the catalytic α subunit of PI3Kα (PIK3CA). These PIK3CA mutations are known to cause solid cancers when occurring in other tissues. PIK3CA-related vascular anomalies, or "PIKopathies," range from simple, i.e., restricted to a particular form of malformation, to complex, i.e., presenting with a range of hyperplasia phenotypes, including the PIK3CA-related overgrowth spectrum. Interestingly, development of PIKopathies is affected by fluid shear stress (FSS), a physiological stimulus caused by blood or lymph flow. These findings implicate PI3K in mediating physiological EC responses to FSS conditions characteristic of lymphatic and capillary vessel beds. Consistent with this hypothesis, increased PI3K signaling also contributes to cerebral cavernous malformations, a vascular disorder that affects low-perfused brain venous capillaries. Because the GOF activity of PI3K and its signaling partners are excellent drug targets, understanding PIK3CA's role in the development of vascular anomalies may inform therapeutic strategies to normalize EC responses in the diseased state. This Review focuses on PIK3CA's role in mediating EC responses to FSS and discusses current understanding of PIK3CA dysregulation in a range of vascular anomalies that particularly affect low-perfused regions of the vasculature. We also discuss recent surprising findings linking increased PI3K signaling to fast-flow arteriovenous malformations in hereditary hemorrhagic telangiectasias.
Collapse
Affiliation(s)
| | - Roxana Ola
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
11
|
Allen-Gondringer A, Gau D, Dutta P, Roy P. Haplo-insufficiency of Profilin1 in vascular endothelial cells is beneficial but not sufficient to confer protection against experimentally induced atherosclerosis. Cytoskeleton (Hoboken) 2024:10.1002/cm.21859. [PMID: 38623956 PMCID: PMC11480255 DOI: 10.1002/cm.21859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
Actin cytoskeleton plays an important role in various aspects of atherosclerosis, a key driver of ischemic heart disease. Actin-binding protein Profilin1 (Pfn1) is overexpressed in atherosclerotic plaques in human disease, and Pfn1, when partially depleted globally in all cell types, confers atheroprotection in vivo. This study investigates the impact of endothelial cell (EC)-specific partial loss of Pfn1 expression in atherosclerosis development. We utilized mice engineered for conditional heterozygous knockout of the Pfn1 gene in ECs, with atherosclerosis induced by depletion of hepatic LDL receptor by gene delivery of PCSK9 combined with high-cholesterol diet. Our studies show that partial depletion of EC Pfn1 has certain beneficial effects marked by dampening of select pro-atherogenic cytokines (CXCL10 and IL7) with concomitant reduction in cytotoxic T cell abundance but is not sufficient to reduce hyperlipidemia and confer atheroprotection in vivo. In light of these findings, we conclude that atheroprotective phenotype conferred by global Pfn1 haplo-insufficiency requires contributions of additional cell types that are relevant for atherosclerosis progression.
Collapse
Affiliation(s)
| | - David Gau
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Partha Dutta
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Partha Roy
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Pathology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
12
|
Lemmens TP, Bröker V, Rijpkema M, Hughes CCW, Schurgers LJ, Cosemans JMEM. Fundamental considerations for designing endothelialized in vitro models of thrombosis. Thromb Res 2024; 236:179-190. [PMID: 38460307 DOI: 10.1016/j.thromres.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Endothelialized in vitro models for cardiovascular disease have contributed greatly to our current understanding of the complex molecular mechanisms underlying thrombosis. To further elucidate these mechanisms, it is important to consider which fundamental aspects to incorporate into an in vitro model. In this review, we will focus on the design of in vitro endothelialized models of thrombosis. Expanding our understanding of the relation and interplay between the different pathways involved will rely in part on complex models that incorporate endothelial cells, blood, the extracellular matrix, and flow. Importantly, the use of tissue-specific endothelial cells will help in understanding the heterogeneity in thrombotic responses between different vascular beds. The dynamic and complex responses of endothelial cells to different shear rates underlines the importance of incorporating appropriate shear in in vitro models. Alterations in vascular extracellular matrix composition, availability of bioactive molecules, and gradients in concentration and composition of these molecules can all regulate the function of both endothelial cells and perivascular cells. Factors modulating these elements in in vitro models should therefore be considered carefully depending on the research question at hand. As the complexity of in vitro models increases, so can the variability. A bottom-up approach to designing such models will remain an important tool for researchers studying thrombosis. As new techniques are continuously being developed and new pathways are brought to light, research question-dependent considerations will have to be made regarding what aspects of thrombosis to include in in vitro models.
Collapse
Affiliation(s)
- Titus P Lemmens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Vanessa Bröker
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Minke Rijpkema
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, and Department of Biomedical Engineering, University of California, Irvine, USA
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
13
|
He J, Blazeski A, Nilanthi U, Menéndez J, Pirani SC, Levic DS, Bagnat M, Singh MK, Raya JG, García-Cardeña G, Torres-Vázquez J. Plxnd1-mediated mechanosensing of blood flow controls the caliber of the Dorsal Aorta via the transcription factor Klf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.576555. [PMID: 38328196 PMCID: PMC10849625 DOI: 10.1101/2024.01.24.576555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The cardiovascular system generates and responds to mechanical forces. The heartbeat pumps blood through a network of vascular tubes, which adjust their caliber in response to the hemodynamic environment. However, how endothelial cells in the developing vascular system integrate inputs from circulatory forces into signaling pathways to define vessel caliber is poorly understood. Using vertebrate embryos and in vitro-assembled microvascular networks of human endothelial cells as models, flow and genetic manipulations, and custom software, we reveal that Plexin-D1, an endothelial Semaphorin receptor critical for angiogenic guidance, employs its mechanosensing activity to serve as a crucial positive regulator of the Dorsal Aorta's (DA) caliber. We also uncover that the flow-responsive transcription factor KLF2 acts as a paramount mechanosensitive effector of Plexin-D1 that enlarges endothelial cells to widen the vessel. These findings illuminate the molecular and cellular mechanisms orchestrating the interplay between cardiovascular development and hemodynamic forces.
Collapse
Affiliation(s)
- Jia He
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Uthayanan Nilanthi
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
| | - Javier Menéndez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Samuel C. Pirani
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel S. Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Manvendra K. Singh
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609
| | - José G Raya
- Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesús Torres-Vázquez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
14
|
Struck EC, Belova T, Hsieh PH, Odeberg JO, Kuijjer ML, Dusart PJ, Butler LM. Global Transcriptome Analysis Reveals Distinct Phases of the Endothelial Response to TNF. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:117-129. [PMID: 38019121 PMCID: PMC10733583 DOI: 10.4049/jimmunol.2300419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/19/2023] [Indexed: 11/30/2023]
Abstract
The vascular endothelium acts as a dynamic interface between blood and tissue. TNF-α, a major regulator of inflammation, induces endothelial cell (EC) transcriptional changes, the overall response dynamics of which have not been fully elucidated. In the present study, we conducted an extended time-course analysis of the human EC response to TNF, from 30 min to 72 h. We identified regulated genes and used weighted gene network correlation analysis to decipher coexpression profiles, uncovering two distinct temporal phases: an acute response (between 1 and 4 h) and a later phase (between 12 and 24 h). Sex-based subset analysis revealed that the response was comparable between female and male cells. Several previously uncharacterized genes were strongly regulated during the acute phase, whereas the majority in the later phase were IFN-stimulated genes. A lack of IFN transcription indicated that this IFN-stimulated gene expression was independent of de novo IFN production. We also observed two groups of genes whose transcription was inhibited by TNF: those that resolved toward baseline levels and those that did not. Our study provides insights into the global dynamics of the EC transcriptional response to TNF, highlighting distinct gene expression patterns during the acute and later phases. Data for all coding and noncoding genes is provided on the Web site (http://www.endothelial-response.org/). These findings may be useful in understanding the role of ECs in inflammation and in developing TNF signaling-targeted therapies.
Collapse
Affiliation(s)
- Eike C. Struck
- Department of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway
| | - Tatiana Belova
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Ping-Han Hsieh
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Jacob O. Odeberg
- Department of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology, Stockholm, Sweden
- The University Hospital of North Norway, Tromsø, Norway
- Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Marieke L. Kuijjer
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Philip J. Dusart
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology, Stockholm, Sweden
- Clinical Chemistry and Blood Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Lynn M. Butler
- Department of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology, Stockholm, Sweden
- Clinical Chemistry and Blood Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
Schumacher JA, Wright ZA, Rufin Florat D, Anand SK, Dasyani M, Batta SPR, Laverde V, Ferrari K, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E and ABL signaling regulate blood vessel size. PLoS Genet 2024; 20:e1010851. [PMID: 38190417 PMCID: PMC10798624 DOI: 10.1371/journal.pgen.1010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/19/2024] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Blood vessels in different vascular beds vary in size, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vessel size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow, eventually leading to the DA collapse. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA size in she mutants correlated with an increased endothelial expression of claudin 5a (cldn5a), which encodes a protein enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates vessel and lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surya Prakash Rao Batta
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Valentina Laverde
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Kaitlin Ferrari
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| |
Collapse
|
16
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
17
|
Allbritton-King JD, García-Cardeña G. Endothelial cell dysfunction in cardiac disease: driver or consequence? Front Cell Dev Biol 2023; 11:1278166. [PMID: 37965580 PMCID: PMC10642230 DOI: 10.3389/fcell.2023.1278166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
The vascular endothelium is a multifunctional cellular system which directly influences blood components and cells within the vessel wall in a given tissue. Importantly, this cellular interface undergoes critical phenotypic changes in response to various biochemical and hemodynamic stimuli, driving several developmental and pathophysiological processes. Multiple studies have indicated a central role of the endothelium in the initiation, progression, and clinical outcomes of cardiac disease. In this review we synthesize the current understanding of endothelial function and dysfunction as mediators of the cardiomyocyte phenotype in the setting of distinct cardiac pathologies; outline existing in vivo and in vitro models where key features of endothelial cell dysfunction can be recapitulated; and discuss future directions for development of endothelium-targeted therapeutics for cardiac diseases with limited existing treatment options.
Collapse
Affiliation(s)
- Jules D. Allbritton-King
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Guillermo García-Cardeña
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
18
|
Bosseboeuf E, Chikh A, Chaker AB, Mitchell TP, Vignaraja D, Rajendrakumar R, Khambata RS, Nightingale TD, Mason JC, Randi AM, Ahluwalia A, Raimondi C. Neuropilin-1 interacts with VE-cadherin and TGFBR2 to stabilize adherens junctions and prevent activation of endothelium under flow. Sci Signal 2023; 16:eabo4863. [PMID: 37220183 PMCID: PMC7614756 DOI: 10.1126/scisignal.abo4863] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-β (TGF-β) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-β signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-β signaling and inflammation.
Collapse
Affiliation(s)
- Emy Bosseboeuf
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Anissa Chikh
- Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London SW17 0RE, UK
| | - Ahmed Bey Chaker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Tom P. Mitchell
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Dhilakshani Vignaraja
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ridhi Rajendrakumar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Rayomand S. Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Thomas D. Nightingale
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Justin C. Mason
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Anna M. Randi
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Claudio Raimondi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
19
|
Abutaleb NO, Atchison L, Choi L, Bedapudi A, Shores K, Gete Y, Cao K, Truskey GA. Lonafarnib and everolimus reduce pathology in iPSC-derived tissue engineered blood vessel model of Hutchinson-Gilford Progeria Syndrome. Sci Rep 2023; 13:5032. [PMID: 36977745 PMCID: PMC10050176 DOI: 10.1038/s41598-023-32035-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, fatal genetic disease that accelerates atherosclerosis. With a limited pool of HGPS patients, clinical trials face unique challenges and require reliable preclinical testing. We previously reported a 3D tissue engineered blood vessel (TEBV) microphysiological system fabricated with iPSC-derived vascular cells from HGPS patients. HGPS TEBVs exhibit features of HGPS atherosclerosis including loss of smooth muscle cells, reduced vasoactivity, excess extracellular matrix (ECM) deposition, inflammatory marker expression, and calcification. We tested the effects of HGPS therapeutics Lonafarnib and Everolimus separately and together, currently in Phase I/II clinical trial, on HGPS TEBVs. Everolimus decreased reactive oxygen species levels, increased proliferation, reduced DNA damage in HGPS vascular cells, and improved vasoconstriction in HGPS TEBVs. Lonafarnib improved shear stress response of HGPS iPSC-derived endothelial cells (viECs) and reduced ECM deposition, inflammation, and calcification in HGPS TEBVs. Combination treatment with Lonafarnib and Everolimus produced additional benefits such as improved endothelial and smooth muscle marker expression and reduced apoptosis, as well as increased TEBV vasoconstriction and vasodilation. These results suggest that a combined trial of both drugs may provide cardiovascular benefits beyond Lonafarnib, if the Everolimus dose can be tolerated.
Collapse
Affiliation(s)
- Nadia O Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leigh Atchison
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leandro Choi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Akhil Bedapudi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kevin Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yantenew Gete
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
20
|
Afshar Y, Ma F, Quach A, Jeong A, Sunshine HL, Freitas V, Jami-Alahmadi Y, Helaers R, Li X, Pellegrini M, Wohlschlegel JA, Romanoski CE, Vikkula M, Iruela-Arispe ML. Transcriptional drifts associated with environmental changes in endothelial cells. eLife 2023; 12:e81370. [PMID: 36971339 PMCID: PMC10168696 DOI: 10.7554/elife.81370] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/26/2023] [Indexed: 03/29/2023] Open
Abstract
Environmental cues, such as physical forces and heterotypic cell interactions play a critical role in cell function, yet their collective contributions to transcriptional changes are unclear. Focusing on human endothelial cells, we performed broad individual sample analysis to identify transcriptional drifts associated with environmental changes that were independent of genetic background. Global gene expression profiling by RNA sequencing and protein expression by liquid chromatography-mass spectrometry directed proteomics distinguished endothelial cells in vivo from genetically matched culture (in vitro) samples. Over 43% of the transcriptome was significantly changed by the in vitro environment. Subjecting cultured cells to long-term shear stress significantly rescued the expression of approximately 17% of genes. Inclusion of heterotypic interactions by co-culture of endothelial cells with smooth muscle cells normalized approximately 9% of the original in vivo signature. We also identified novel flow dependent genes, as well as genes that necessitate heterotypic cell interactions to mimic the in vivo transcriptome. Our findings highlight specific genes and pathways that rely on contextual information for adequate expression from those that are agnostic of such environmental cues.
Collapse
Affiliation(s)
- Yalda Afshar
- Department of Obstetrics and Gynecology, University of California, Los AngelesLos AngelesUnited States
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
| | - Feyiang Ma
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Austin Quach
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Anhyo Jeong
- Department of Obstetrics and Gynecology, University of California, Los AngelesLos AngelesUnited States
| | - Hannah L Sunshine
- Department of Molecular, Cellular and Integrative Physiology, University of California, Los AngelesLos AngelesUnited States
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Vanessa Freitas
- Departament of Cell and Developmental Biology, Institute of Biomedical Science, University of Sao PauloLos AngelesUnited States
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of CaliforniaLos AngelesUnited States
| | - Raphael Helaers
- Human Molecular Genetics, de Duve Institute, University of LouvainBrusselsBelgium
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos AngelesUnited States
| | - Matteo Pellegrini
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of CaliforniaLos AngelesUnited States
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of LouvainBrusselsBelgium
- WELBIO department, WEL Research InstituteWavreBelgium
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of MedicineChicagoUnited States
| |
Collapse
|
21
|
Huang H, Ren P, Zhao Y, Weng H, Jia C, Yu F, Nie Y. Low shear stress induces inflammatory response via CX3CR1/NF-κB signal pathway in human umbilical vein endothelial cells. Tissue Cell 2023; 82:102043. [PMID: 36827822 DOI: 10.1016/j.tice.2023.102043] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Low shear stress (LSS) has been reported to induce atherosclerosis. However, the molecular mechanisms underlying inflammation induced by LSS are still poorly understood. The objective of our study is the comprehensive identification of molecular circuitry involved in low shear stress-induced inflammation in human umbilical vein endothelial cells (HUVECs) through protein profiling and cell function experiment. In this study, Western blotting analyses revealed a significant increase in the expression of CX3CR1, nucleusP65, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and Interleukin-6 (IL-6), while the expression of cytosolic P65 and IκB has significantly decreased in HUVECs treated with low shear stress. CX3CR1 Sh-RNA was use to reveal its effect on LSS-induced inflammation. Further, specific NF-κB P65 inhibitors pyrrolidinedithiocarbamate (PDTC) were used to reveal the downstream NF-κB P65 exclusively involved in LSS-induced inflammation in HUVECs, this effect can be abrogated by CX3CR1 sh-RNA and NF-κB inhibitors. Monocyte adhesion assay and scratch test revealed low shear stress to promotes adhesion of monocytes and migration of cells, this effect can be abrogated by CX3CR1 sh-RNA and NF-κB inhibitors. LSS was involved in the expression of adhesion molecules and chemokines, which are important for the initiation of endothelial inflammation-related atherosclerosis. Therefore, the cell signaling pathways activated by LSS in endothelial cells may represent therapeutic targets of atherosclerosis.
Collapse
Affiliation(s)
- Haozhong Huang
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Peile Ren
- Department of Physiology, School of Medicine, Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China.
| | - Yiwei Zhao
- The School of Medicine, Huanghe S&T College, Zhengzhou 450063, Henan, PR China.
| | - Huimin Weng
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Chunsen Jia
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Fengxu Yu
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China; Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, PR China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, PR China.
| | - Yongmei Nie
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China; Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, PR China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, PR China.
| |
Collapse
|
22
|
Lin S, Lin R, Zhang H, Xu Q, He Y. Peripheral vascular remodeling during ischemia. Front Pharmacol 2022; 13:1078047. [DOI: 10.3389/fphar.2022.1078047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
About 230 million people worldwide suffer from peripheral arterial disease (PAD), and the prevalence is increasing year by year. Multiple risk factors, including smoking, dyslipidemia, diabetes, and hypertension, can contribute to the development of PAD. PAD is typically characterized by intermittent claudication and resting pain, and there is a risk of severe limb ischemia, leading to major adverse limb events, such as amputation. Currently, a major progress in the research field of the pathogenesis of vascular remodeling, including atherosclerosis and neointima hyperplasia has been made. For example, the molecular mechanisms of endothelial dysfunction and smooth muscle phenotype switching have been described. Interestingly, a series of focused studies on fibroblasts of the vessel wall has demonstrated their impact on smooth muscle proliferation and even endothelial function via cell-cell communications. In this review, we aim to focus on the functional changes of peripheral arterial cells and the mechanisms of the pathogenesis of PAD. At the same time, we summarize the progress of the current clinical treatment and potential therapeutic methods for PAD and shine a light on future perspectives.
Collapse
|
23
|
Weaver SRC, Rendeiro C, Lucas RAI, Cable NT, Nightingale TE, McGettrick HM, Lucas SJE. Non-pharmacological interventions for vascular health and the role of the endothelium. Eur J Appl Physiol 2022. [PMID: 36149520 DOI: 10.1007/s00421-022-05041-y.pmid:36149520;pmcid:pmc9613570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
The most common non-pharmacological intervention for both peripheral and cerebral vascular health is regular physical activity (e.g., exercise training), which improves function across a range of exercise intensities and modalities. Numerous non-exercising approaches have also been suggested to improved vascular function, including repeated ischemic preconditioning (IPC); heat therapy such as hot water bathing and sauna; and pneumatic compression. Chronic adaptive responses have been observed across a number of these approaches, yet the precise mechanisms that underlie these effects in humans are not fully understood. Acute increases in blood flow and circulating signalling factors that induce responses in endothelial function are likely to be key moderators driving these adaptations. While the impact on circulating factors and environmental mechanisms for adaptation may vary between approaches, in essence, they all centre around acutely elevating blood flow throughout the circulation and stimulating improved endothelium-dependent vascular function and ultimately vascular health. Here, we review our current understanding of the mechanisms driving endothelial adaptation to repeated exposure to elevated blood flow, and the interplay between this response and changes in circulating factors. In addition, we will consider the limitations in our current knowledge base and how these may be best addressed through the selection of more physiologically relevant experimental models and research. Ultimately, improving our understanding of the unique impact that non-pharmacological interventions have on the vasculature will allow us to develop superior strategies to tackle declining vascular function across the lifespan, prevent avoidable vascular-related disease, and alleviate dependency on drug-based interventions.
Collapse
Affiliation(s)
- Samuel R C Weaver
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK.
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK.
| | - Catarina Rendeiro
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Rebekah A I Lucas
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - N Timothy Cable
- Institute of Sport, Manchester Metropolitan University, Manchester, UK
| | - Tom E Nightingale
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Samuel J E Lucas
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
24
|
Fernandes A, Miéville A, Grob F, Yamashita T, Mehl J, Hosseini V, Emmert MY, Falk V, Vogel V. Endothelial-Smooth Muscle Cell Interactions in a Shear-Exposed Intimal Hyperplasia on-a-Dish Model to Evaluate Therapeutic Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202317. [PMID: 35971167 PMCID: PMC9534971 DOI: 10.1002/advs.202202317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Indexed: 05/25/2023]
Abstract
Intimal hyperplasia (IH) represents a major challenge following cardiovascular interventions. While mechanisms are poorly understood, the inefficient preventive methods incentivize the search for novel therapies. A vessel-on-a-dish platform is presented, consisting of direct-contact cocultures with human primary endothelial cells (ECs) and smooth muscle cells (SMCs) exposed to both laminar pulsatile and disturbed flow on an orbital shaker. With contractile SMCs sitting below a confluent EC layer, a model that successfully replicates the architecture of a quiescent vessel wall is created. In the novel IH model, ECs are seeded on synthetic SMCs at low density, mimicking reendothelization after vascular injury. Over 3 days of coculture, ECs transition from a network conformation to confluent 2D islands, as promoted by pulsatile flow, resulting in a "defected" EC monolayer. In defected regions, SMCs incorporated plasma fibronectin into fibers, increased proliferation, and formed multilayers, similarly to IH in vivo. These phenomena are inhibited under confluent EC layers, supporting therapeutic approaches that focus on endothelial regeneration rather than inhibiting proliferation, as illustrated in a proof-of-concept experiment with Paclitaxel. Thus, this in vitro system offers a new tool to study EC-SMC communication in IH pathophysiology, while providing an easy-to-use translational disease model platform for low-cost and high-content therapeutic development.
Collapse
Affiliation(s)
- Andreia Fernandes
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Arnaud Miéville
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Franziska Grob
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Tadahiro Yamashita
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
- Present address:
Department of System Design EngineeringKeio University108‐8345YokohamaJapan
| | - Julia Mehl
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
- Present address:
Julius Wolff InstituteBerlin Institute of HealthCharité Universitätsmedizin Berlin10117BerlinGermany
| | - Vahid Hosseini
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Maximilian Y. Emmert
- Department of Cardiovascular SurgeryCharité Universitätsmedizin Berlin10117BerlinGermany
- Department of Cardiothoracic and Vascular SurgeryGerman Heart Center Berlin13353BerlinGermany
- Institute for Regenerative Medicine (IREM)University of Zurich8006ZurichSwitzerland
| | - Volkmar Falk
- Department of Cardiovascular SurgeryCharité Universitätsmedizin Berlin10117BerlinGermany
- Department of Cardiothoracic and Vascular SurgeryGerman Heart Center Berlin13353BerlinGermany
- Department of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Viola Vogel
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| |
Collapse
|
25
|
Weaver SRC, Rendeiro C, Lucas RAI, Cable NT, Nightingale TE, McGettrick HM, Lucas SJE. Non-pharmacological interventions for vascular health and the role of the endothelium. Eur J Appl Physiol 2022; 122:2493-2514. [PMID: 36149520 PMCID: PMC9613570 DOI: 10.1007/s00421-022-05041-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/05/2022] [Indexed: 12/11/2022]
Abstract
The most common non-pharmacological intervention for both peripheral and cerebral vascular health is regular physical activity (e.g., exercise training), which improves function across a range of exercise intensities and modalities. Numerous non-exercising approaches have also been suggested to improved vascular function, including repeated ischemic preconditioning (IPC); heat therapy such as hot water bathing and sauna; and pneumatic compression. Chronic adaptive responses have been observed across a number of these approaches, yet the precise mechanisms that underlie these effects in humans are not fully understood. Acute increases in blood flow and circulating signalling factors that induce responses in endothelial function are likely to be key moderators driving these adaptations. While the impact on circulating factors and environmental mechanisms for adaptation may vary between approaches, in essence, they all centre around acutely elevating blood flow throughout the circulation and stimulating improved endothelium-dependent vascular function and ultimately vascular health. Here, we review our current understanding of the mechanisms driving endothelial adaptation to repeated exposure to elevated blood flow, and the interplay between this response and changes in circulating factors. In addition, we will consider the limitations in our current knowledge base and how these may be best addressed through the selection of more physiologically relevant experimental models and research. Ultimately, improving our understanding of the unique impact that non-pharmacological interventions have on the vasculature will allow us to develop superior strategies to tackle declining vascular function across the lifespan, prevent avoidable vascular-related disease, and alleviate dependency on drug-based interventions.
Collapse
Affiliation(s)
- Samuel R C Weaver
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK.
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK.
| | - Catarina Rendeiro
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Rebekah A I Lucas
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - N Timothy Cable
- Institute of Sport, Manchester Metropolitan University, Manchester, UK
| | - Tom E Nightingale
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Samuel J E Lucas
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
26
|
On non-Kolmogorov turbulence in blood flow and its possible role in mechanobiological stimulation. Sci Rep 2022; 12:13166. [PMID: 35915207 PMCID: PMC9343407 DOI: 10.1038/s41598-022-16079-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/04/2022] [Indexed: 01/09/2023] Open
Abstract
The study of turbulence in physiologic blood flow is important due to its strong relevance to endothelial mechanobiology and vascular disease. Recently, Saqr et al. (Sci Rep 10, 15,492, 2020) discovered non-Kolmogorov turbulence in physiologic blood flow in vivo, traced its origins to the Navier–Stokes equation and demonstrated some of its properties using chaos and hydrodynamic-stability theories. The present work extends these findings and investigates some inherent characteristics of non-Kolmogorov turbulence in monoharmonic and multiharmonic pulsatile flow under ideal physiologic conditions. The purpose of this work is to propose a conjecture for the origins for picoNewton forces that are known to regulate endothelial cells’ functions. The new conjecture relates these forces to physiologic momentum-viscous interactions in the near-wall region of the flow. Here, we used high-resolution large eddy simulation (HRLES) to study pulsatile incompressible flow in a straight pipe of \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$L/D=20$$\end{document}L/D=20. The simulations presented Newtonian and Carreau–Yasuda fluid flows, at \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$R{e}_{m}\approx 250$$\end{document}Rem≈250, each represented by one, two and three boundary harmonics. Comparison was established based on maintaining constant time-averaged mass flow rate in all simulations. First, we report the effect of primary harmonics on the global power budget using primitive variables in phase space. Second, we describe the non-Kolmogorov turbulence in frequency domain. Third, we investigate the near-wall coherent structures in time and space domains. Finally, we propose a new conjecture for the role of turbulence in endothelial cells’ mechanobiology. The proposed conjecture correlates near-wall turbulence to a force field of picoNewton scale, suggesting possible relevance to endothelial cells mechanobiology.
Collapse
|
27
|
Deng H, Schwartz MA. High Fluid Shear Stress Inhibits Cytokine-Driven Smad2/3 Activation in Vascular Endothelial Cells. J Am Heart Assoc 2022; 11:e025337. [PMID: 35861829 PMCID: PMC9707828 DOI: 10.1161/jaha.121.025337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/16/2022] [Indexed: 01/22/2023]
Abstract
Background Atherosclerosis occurs preferentially in regions of low and disturbed fluid shear stress (FSS) but is limited in regions of high laminar FSS as a result of inhibition of endothelial inflammatory pathways. Recent work has identified endothelial to mesenchymal transition (EndMT) driven by TGFβ2 (transforming growth factor beta 2)-Smad2/3 (mothers against decapentaplegic) signaling as a critical component of atherogenesis. However, interactions between FSS and EndMT in this context have not been investigated. Methods and Results Endothelial cells were treated with TGFβ2 and inflammatory cytokines (interleukin 1β and tumor necrosis factor alpha) with or without high FSS in a parallel plate flow chamber. Smad2/3 nuclear translocation and target gene expression, assayed by immunofluorescence and quantitative polymerase chain reaction, revealed that high FSS blocked the Smad2/3-EndMT pathway. In vivo, mice were injected with TGFβ2 and inflammatory cytokines, then regions of the aorta under low versus high FSS were examined. TGFβ2 and inflammatory cytokine treatment stimulated Smad2/3 nuclear translocation and target gene expression predominantly in regions of low FSS with little effect in regions of high FSS. Conclusions High FSS inhibits endothelial Smad2/3 activation and EndMT in response to inflammatory mediators, resulting in selective EndMT at athero-susceptible, low FSS regions of arteries.
Collapse
Affiliation(s)
- Hanqiang Deng
- Department of Internal Medicine, Yale Cardiovascular Research CenterYale University School of MedicineNew HavenCT
| | - Martin A. Schwartz
- Department of Internal Medicine, Yale Cardiovascular Research CenterYale University School of MedicineNew HavenCT
- Department of Cell BiologyYale University School of MedicineNew HavenCT
- Department of Biomedical EngineeringYale UniversityNew HavenCT
| |
Collapse
|
28
|
Dupont S, Wickström SA. Mechanical regulation of chromatin and transcription. Nat Rev Genet 2022; 23:624-643. [DOI: 10.1038/s41576-022-00493-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 01/14/2023]
|
29
|
Kutikhin AG, Shishkova DK, Velikanova EA, Sinitsky MY, Sinitskaya AV, Markova VE. Endothelial Dysfunction in the Context of Blood–Brain Barrier Modeling. J EVOL BIOCHEM PHYS+ 2022; 58:781-806. [PMID: 35789679 PMCID: PMC9243926 DOI: 10.1134/s0022093022030139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023]
Abstract
Here, we discuss pathophysiological approaches to the defining
of endothelial dysfunction criteria (i.e., endothelial activation,
impaired endothelial mechanotransduction, endothelial-to-mesenchymal
transition, reduced nitric oxide release, compromised endothelial
integrity, and loss of anti-thrombogenic properties) in different
in vitro and in vivo models. The canonical definition of endothelial
dysfunction includes insufficient production of vasodilators, pro-thrombotic
and pro-inflammatory activation of endothelial cells, and pathologically
increased endothelial permeability. Among the clinical consequences
of endothelial dysfunction are arterial hypertension, macro- and
microangiopathy, and microalbuminuria. We propose to extend the definition
of endothelial dysfunction by adding altered endothelial mechanotransduction
and endothelial-to-mesenchymal transition to its criteria. Albeit
interleukin-6, interleukin-8, and MCP-1/CCL2 dictate the pathogenic
paracrine effects of dysfunctional endothelial cells and are therefore
reliable endothelial dysfunction biomarkers in vitro, they are non-specific
for endothelial cells and cannot be used for the diagnostics of
endothelial dysfunction in vivo. Conceptual improvements in the
existing methods to model endothelial dysfunction, specifically,
in relation to the blood–brain barrier, include endothelial cell
culturing under pulsatile flow, collagen IV coating of flow chambers,
and endothelial lysate collection from the blood vessels of laboratory
animals in situ for the subsequent gene and protein expression profiling.
Combined with the simulation of paracrine effects by using conditioned
medium from dysfunctional endothelial cells, these flow-sensitive
models have a high physiological relevance, bringing the experimental
conditions to the physiological scenario.
Collapse
Affiliation(s)
- A. G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - D. K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - E. A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - M. Yu. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A. V. Sinitskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - V. E. Markova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
30
|
Liang G, Wang S, Shao J, Jin Y, Xu L, Yan Y, Günther S, Wang L, Offermanns S. Tenascin-X Mediates Flow-Induced Suppression of EndMT and Atherosclerosis. Circ Res 2022; 130:1647-1659. [PMID: 35443807 DOI: 10.1161/circresaha.121.320694] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial-to-mesenchymal transition (EndMT) has been identified as a critical driver of vascular inflammation and atherosclerosis, and TGF-β (transforming growth factor β) is a key mediator of EndMT. Both EndMT and atherosclerosis are promoted by disturbed flow, whereas unidirectional laminar flow limits EndMT and is atheroprotective. How EndMT and endothelial TGF-β signaling are regulated by different flow patterns is, however, still poorly understood. METHODS Flow chamber experiments in vitro and endothelium-specific knockout mice were used to study the role of tenascin-X in the regulation of EndMT and atherosclerosis as well as the underlying mechanisms. RESULTS In human endothelial cells as well as in human and mouse aortae, unidirectional laminar flow but not disturbed flow strongly increased endothelial expression of the extracellular matrix protein TN-X (tenascin-X) in a KLF4 (Krüppel-like factor 4) dependent manner. Mice with endothelium-specific loss of TN-X (EC-Tnxb-KO) showed increased endothelial TGF-β signaling as well as increased endothelial expression of EndMT and inflammatory marker genes. When EC-Tnxb-KO mice were subjected to partial carotid artery ligation, we observed increased vascular remodeling. EC-Tnxb-KO mice crossed to low-density lipoprotein receptor-deficient mice showed advanced atherosclerotic lesions after being fed a high-fat diet. Treatment of EC-Tnxb-KO mice with an anti-TGF-beta antibody or additional endothelial loss of TGF-beta receptors 1 and 2 normalized endothelial TGF-beta signaling and prevented EndMT. In in vitro studies, we found that TN-X through its fibrinogen-like domain directly interacts with TGF-β and thereby interferes with its binding to the TGF-β receptor. CONCLUSIONS In summary, we show that TN-X is a central mediator of flow-induced inhibition of EndMT, endothelial inflammation and atherogenesis, which functions by binding to and by blocking the activity of TGF-β. Our data identify a novel mechanism of flow-dependent regulation of vascular TGF-β, which holds promise for generating new strategies to prevent vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - ShengPeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (S.W., L.X.)
| | - Jingchen Shao
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - YoungJune Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - Liran Xu
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (S.W., L.X.)
| | - Yang Yan
- Department of Cardiovascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, China (Y.Y.)
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Germany (S.G.)
| | - Lei Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.).,Center for Molecular Medicine, Goethe University Frankfurt, Germany (S.O.).,Cardiopulmonary Institute (CPI), Frankfurt/Bad Nauheim, Germany (S.O.).,German Center for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany (S.O.)
| |
Collapse
|
31
|
Tsaryk R, Yucel N, Leonard EV, Diaz N, Bondareva O, Odenthal-Schnittler M, Arany Z, Vaquerizas JM, Schnittler H, Siekmann AF. Shear stress switches the association of endothelial enhancers from ETV/ETS to KLF transcription factor binding sites. Sci Rep 2022; 12:4795. [PMID: 35314737 PMCID: PMC8938417 DOI: 10.1038/s41598-022-08645-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) lining blood vessels are exposed to mechanical forces, such as shear stress. These forces control many aspects of EC biology, including vascular tone, cell migration and proliferation. Despite a good understanding of the genes responding to shear stress, our insight into the transcriptional regulation of these genes is much more limited. Here, we set out to study alterations in the chromatin landscape of human umbilical vein endothelial cells (HUVEC) exposed to laminar shear stress. To do so, we performed ChIP-Seq for H3K27 acetylation, indicative of active enhancer elements and ATAC-Seq to mark regions of open chromatin in addition to RNA-Seq on HUVEC exposed to 6 h of laminar shear stress. Our results show a correlation of gained and lost enhancers with up and downregulated genes, respectively. DNA motif analysis revealed an over-representation of KLF transcription factor (TF) binding sites in gained enhancers, while lost enhancers contained more ETV/ETS motifs. We validated a subset of flow responsive enhancers using luciferase-based reporter constructs and CRISPR-Cas9 mediated genome editing. Lastly, we characterized the shear stress response in ECs of zebrafish embryos using RNA-Seq. Our results lay the groundwork for the exploration of shear stress responsive elements in controlling EC biology.
Collapse
Affiliation(s)
- Roman Tsaryk
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nora Yucel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Elvin V Leonard
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Noelia Diaz
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Olga Bondareva
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103, Leipzig, Germany
| | - Maria Odenthal-Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Juan M Vaquerizas
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Hans Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany.
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany.
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Sigler M, Rouatbi H, Vazquez-Jimenez J, Seghaye MC. Uni-ventricular palliation vs. bi-ventricular repair: differential inflammatory response. Mol Cell Pediatr 2022; 9:5. [PMID: 35307783 PMCID: PMC8934903 DOI: 10.1186/s40348-022-00138-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
Background To examine whether uni-ventricular palliation (UVP) and bi-ventricular repair (BVR) result in a different pattern of systemic inflammatory response to pediatric cardiac surgery with extra-corporeal circulation (ECC). Methods In 20 children (median age 39.5 months) undergoing either UVP (n = 12) or BVR (n = 8), plasma levels of the inflammatory cytokines TNF-α, IL-6, IL-10, and IL-12 and of procalcitonin (PCT), were measured before, during and after open cardiac surgery up to postoperative day (POD) 10. Results Epidemiologic, operative- and outcome variables were similar in both groups but post-operative central venous pressure that was higher in UVP. In the whole cohort, the inflammatory response was characterized by an early important, significant and parallel increase of IL-6 and IL-10 that reached their peak values either at the end of ECC (IL-10) or 4 h postoperatively (IL-6), respectively and by a significant and parallel decrease of TNF-α and IL-12 levels after connection to ECC, followed by a bi-phasic significant increase with a first peak 4 h after ECC and a second at POD 10, respectively. Patients after UVP showed a shift of the cytokine balance with lower IL-6- (p = 0.01) after connection to ECC, lower early post-operative TNF-α - (p = 0.02) and IL-12- (p = 0.04) concentrations and lower TNF-α/IL-10-ratio (p = 0.03) as compared with patients with BVR. Levels of PCT were similar in both groups. Conclusions UVP is associated with an anti-inflammatory shift of the inflammatory response to cardiac surgery that might be related to the particular hemodynamic situation of patients with UVP.
Collapse
Affiliation(s)
- Matthias Sigler
- Pediatric Cardiology, Intensive Care Medicine and Neonatology, Georg-August Universität, Robert-Koch-Str. 40, D-37075, Göttingen, Germany.
| | - Hatem Rouatbi
- Department of Pediatrics and Pediatric Cardiology, University Hospital Liège, Liège, Belgium
| | - Jaime Vazquez-Jimenez
- Department of Pediatric Cardiac Surgery, University Hospital Aachen, Aachen, Germany
| | - Marie-Christine Seghaye
- Department of Pediatrics and Pediatric Cardiology, University Hospital Liège, Liège, Belgium
| |
Collapse
|
33
|
Sunderland K, Jiang J, Zhao F. Disturbed flow's impact on cellular changes indicative of vascular aneurysm initiation, expansion, and rupture: A pathological and methodological review. J Cell Physiol 2022; 237:278-300. [PMID: 34486114 PMCID: PMC8810685 DOI: 10.1002/jcp.30569] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Aneurysms are malformations within the arterial vasculature brought on by the structural breakdown of the microarchitecture of the vessel wall, with aneurysms posing serious health risks in the event of their rupture. Blood flow within vessels is generally laminar with high, unidirectional wall shear stressors that modulate vascular endothelial cell functionality and regulate vascular smooth muscle cells. However, altered vascular geometry induced by bifurcations, significant curvature, stenosis, or clinical interventions can alter the flow, generating low stressor disturbed flow patterns. Disturbed flow is associated with altered cellular morphology, upregulated expression of proteins modulating inflammation, decreased regulation of vascular permeability, degraded extracellular matrix, and heightened cellular apoptosis. The understanding of the effects disturbed flow has on the cellular cascades which initiate aneurysms and promote their subsequent growth can further elucidate the nature of this complex pathology. This review summarizes the current knowledge about the disturbed flow and its relation to aneurysm pathology, the methods used to investigate these relations, as well as how such knowledge has impacted clinical treatment methodologies. This information can contribute to the understanding of the development, growth, and rupture of aneurysms and help develop novel research and aneurysmal treatment techniques.
Collapse
Affiliation(s)
- Kevin Sunderland
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Jingfeng Jiang
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| |
Collapse
|
34
|
Lai A, Cox CD, Chandra Sekar N, Thurgood P, Jaworowski A, Peter K, Baratchi S. Mechanosensing by Piezo1 and its implications for physiology and various pathologies. Biol Rev Camb Philos Soc 2021; 97:604-614. [PMID: 34781417 DOI: 10.1111/brv.12814] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/21/2021] [Accepted: 11/04/2021] [Indexed: 11/27/2022]
Abstract
Piezo1 is a mechanosensitive ion channel with essential roles in cardiovascular, lung, urinary, and immune functions. Piezo1 is widely distributed in different tissues in the human body and its specific roles have been identified following a decade of research; however, not all are well understood. Many structural and functional characteristics of Piezo1 have been discovered and are known to differ greatly from the characteristics of other mechanosensitive ion channels. Understanding the mechanisms by which this ion channel functions may be useful in determining its physiological roles in various organ systems. This review provides insight into the signalling pathways activated by mechanical stimulation of Piezo1 in various organ systems and cell types. We discuss downstream targets of Piezo1 and the overall effects resulting from Piezo1 activation, which may provide insights into potential treatment targets for diseases involving this ion channel.
Collapse
Affiliation(s)
- Austin Lai
- School of Health and Biomedical Sciences, RMIT University, 289 McKimmies Rd, Bundoora, Victoria, 3083, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, 405 Liverpool St, Sydney, New South Wales, 2010, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, 289 McKimmies Rd, Bundoora, Victoria, 3083, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, 124 La Trobe St, Melbourne, Victoria, 3001, Australia
| | - Anthony Jaworowski
- School of Health and Biomedical Sciences, RMIT University, 289 McKimmies Rd, Bundoora, Victoria, 3083, Australia
| | - Karlheinz Peter
- School of Health and Biomedical Sciences, RMIT University, 289 McKimmies Rd, Bundoora, Victoria, 3083, Australia.,Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, Victoria, 3004, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, 30 Flemington Rd, Parkville, 3053, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, 289 McKimmies Rd, Bundoora, Victoria, 3083, Australia.,Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, Victoria, 3004, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, 30 Flemington Rd, Parkville, 3053, Australia
| |
Collapse
|
35
|
Castle EL, Robinson CA, Douglas P, Rinker KD, Corcoran JA. Viral Manipulation of a Mechanoresponsive Signaling Axis Disassembles Processing Bodies. Mol Cell Biol 2021; 41:e0039921. [PMID: 34516278 PMCID: PMC8547432 DOI: 10.1128/mcb.00399-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/28/2021] [Accepted: 09/04/2021] [Indexed: 12/23/2022] Open
Abstract
Processing bodies (PBs) are ribonucleoprotein granules important for cytokine mRNA decay that are targeted for disassembly by many viruses. Kaposi's sarcoma-associated herpesvirus is the etiological agent of the inflammatory endothelial cancer, Kaposi's sarcoma, and a PB-regulating virus. The virus encodes kaposin B (KapB), which induces actin stress fibers (SFs) and cell spindling as well as PB disassembly. We now show that KapB-mediated PB disassembly requires actin rearrangements, RhoA effectors, and the mechanoresponsive transcription activator, YAP. Moreover, ectopic expression of active YAP or exposure of ECs to mechanical forces caused PB disassembly in the absence of KapB. We propose that the viral protein KapB activates a mechanoresponsive signaling axis and links changes in cell shape and cytoskeletal structures to enhanced inflammatory molecule expression using PB disassembly. Our work implies that cytoskeletal changes in other pathologies may similarly impact the inflammatory environment.
Collapse
Affiliation(s)
- Elizabeth L. Castle
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolyn-Ann Robinson
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Pauline Douglas
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kristina D. Rinker
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Chemical and Petroleum Engineering and Centre for Bioengineering Research and Education, University of Calgary, Calgary, Alberta, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer A. Corcoran
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
36
|
Parra-Izquierdo I, Sánchez-Bayuela T, López J, Gómez C, Pérez-Riesgo E, San Román JA, Sánchez Crespo M, Yacoub M, Chester AH, García-Rodríguez C. Interferons Are Pro-Inflammatory Cytokines in Sheared-Stressed Human Aortic Valve Endothelial Cells. Int J Mol Sci 2021; 22:ijms221910605. [PMID: 34638942 PMCID: PMC8508640 DOI: 10.3390/ijms221910605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is an athero-inflammatory process. Growing evidence supports the inflammation-driven calcification model, mediated by cytokines such as interferons (IFNs) and tumor necrosis factor (TNF)-α. Our goal was investigating IFNs' effects in human aortic valve endothelial cells (VEC) and the potential differences between aortic (aVEC) and ventricular (vVEC) side cells. The endothelial phenotype was analyzed by Western blot, qPCR, ELISA, monocyte adhesion, and migration assays. In mixed VEC populations, IFNs promoted the activation of signal transducers and activators of transcription-1 and nuclear factor-κB, and the subsequent up-regulation of pro-inflammatory molecules. Side-specific VEC were activated with IFN-γ and TNF-α in an orbital shaker flow system. TNF-α, but not IFN-γ, induced hypoxia-inducible factor (HIF)-1α stabilization or endothelial nitric oxide synthase downregulation. Additionally, IFN-γ inhibited TNF-α-induced migration of aVEC. Also, IFN-γ triggered cytokine secretion and adhesion molecule expression in aVEC and vVEC. Finally, aVEC were more prone to cytokine-mediated monocyte adhesion under multiaxial flow conditions as compared with uniaxial flow. In conclusion, IFNs promote inflammation and reduce TNF-α-mediated migration in human VEC. Moreover, monocyte adhesion was higher in inflamed aVEC sheared under multiaxial flow, which may be relevant to understanding the initial stages of CAVD.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Tania Sánchez-Bayuela
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Javier López
- ICICOR, Hospital Clínico Universitario, 47005 Valladolid, Spain; (J.L.); (J.A.S.R.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Cristina Gómez
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Enrique Pérez-Riesgo
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - J. Alberto San Román
- ICICOR, Hospital Clínico Universitario, 47005 Valladolid, Spain; (J.L.); (J.A.S.R.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Magdi Yacoub
- National Heart & Lung Institute, Imperial College London, London SW3 6LR, UK;
- Magdi Yacoub Institute, Harefield UB9 6JH, UK
| | - Adrian H. Chester
- National Heart & Lung Institute, Imperial College London, London SW3 6LR, UK;
- Magdi Yacoub Institute, Harefield UB9 6JH, UK
- Correspondence: (A.H.C.); (C.G.-R.); Tel.: +44-(0)1895-760732 (A.H.C.); +34-983-184841 (C.G.-R.)
| | - Carmen García-Rodríguez
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: (A.H.C.); (C.G.-R.); Tel.: +44-(0)1895-760732 (A.H.C.); +34-983-184841 (C.G.-R.)
| |
Collapse
|
37
|
Chen ZB, Liu X, Chen AT. "Enhancing" mechanosensing: Enhancers and enhancer-derived long non-coding RNAs in endothelial response to flow. CURRENT TOPICS IN MEMBRANES 2021; 87:153-169. [PMID: 34696884 DOI: 10.1016/bs.ctm.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Endothelial cells (ECs), uniquely localized and strategically forming the inner lining of vascular wall, constitute the largest cell surface by area in the human body. The dynamic sensing and response of ECs to mechanical cues, especially shear stress, is crucial for maintenance of vascular homeostasis. It is well recognized that different flow patterns associated with atheroprotective vs atheroprone regions in the arterial tree, result in distinct EC functional phenotypes with differential transcriptome profiles. Mounting evidence has demonstrated an integrative and essential regulatory role of non-coding genome in EC biology. In particular, recent studies have begun to reveal the importance of enhancers and enhancer-derived transcripts in flow-regulated EC gene expression and function. In this minireview, we summarize studies in this area and discuss examples in support of the emerging importance of enhancers and enhancer(-derived) long non-coding RNAs (elncRNAs) in EC mechanosensing, with a focus on flow-responsive EC transcription. Finally, we will provide perspective and discuss standing questions to elucidate the role of these novel regulators in EC mechanobiology.
Collapse
Affiliation(s)
- Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Duarte, CA, United States; Irell and Manella Graduate School of Biological Sciences, Duarte, CA, United States.
| | - Xuejing Liu
- Department of Diabetes Complications and Metabolism, Duarte, CA, United States
| | - Aleysha T Chen
- Department of Bioengineering, University of California, Berkeley, CA, United States
| |
Collapse
|
38
|
Deng H, Min E, Baeyens N, Coon BG, Hu R, Zhuang ZW, Chen M, Huang B, Afolabi T, Zarkada G, Acheampong A, McEntee K, Eichmann A, Liu F, Su B, Simons M, Schwartz MA. Activation of Smad2/3 signaling by low fluid shear stress mediates artery inward remodeling. Proc Natl Acad Sci U S A 2021; 118:e2105339118. [PMID: 34504019 PMCID: PMC8449390 DOI: 10.1073/pnas.2105339118] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 11/18/2022] Open
Abstract
Endothelial cell (EC) sensing of wall fluid shear stress (FSS) from blood flow governs vessel remodeling to maintain FSS at a specific magnitude or set point in healthy vessels. Low FSS triggers inward remodeling to restore normal FSS but the regulatory mechanisms are unknown. In this paper, we describe the signaling network that governs inward artery remodeling. FSS induces Smad2/3 phosphorylation through the type I transforming growth factor (TGF)-β family receptor Alk5 and the transmembrane protein Neuropilin-1, which together increase sensitivity to circulating bone morphogenetic protein (BMP)-9. Smad2/3 nuclear translocation and target gene expression but not phosphorylation are maximal at low FSS and suppressed at physiological high shear. Reducing flow by carotid ligation in rodents increases Smad2/3 nuclear localization, while the resultant inward remodeling is blocked by the EC-specific deletion of Alk5. The flow-activated MEKK3/Klf2 pathway mediates the suppression of Smad2/3 nuclear translocation at high FSS, mainly through the cyclin-dependent kinase (CDK)-2-dependent phosphosphorylation of the Smad linker region. Thus, low FSS activates Smad2/3, while higher FSS blocks nuclear translocation to induce inward artery remodeling, specifically at low FSS. These results are likely relevant to inward remodeling in atherosclerotic vessels, in which Smad2/3 is activated through TGF-β signaling.
Collapse
Affiliation(s)
- Hanqiang Deng
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Elizabeth Min
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06511
| | - Nicolas Baeyens
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511;
- Department of Internal Medicine (Cardiology), Yale School of Medicine, New Haven, CT 06511
- Laboratoire de Physiologie et Pharmacologie, Faculty of Medicine, Université libre de Bruxelles, 1050 Bruxelles, Belgium
| | - Brian G Coon
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Rui Hu
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Zhen W Zhuang
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
- Department of Physiology, Yale School of Medicine, New Haven, CT 06511
| | - Minghao Chen
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Billy Huang
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Titilayo Afolabi
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Georgia Zarkada
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
| | - Angela Acheampong
- Laboratoire de Physiologie et Pharmacologie, Faculty of Medicine, Université libre de Bruxelles, 1050 Bruxelles, Belgium
| | - Kathleen McEntee
- Laboratoire de Physiologie et Pharmacologie, Faculty of Medicine, Université libre de Bruxelles, 1050 Bruxelles, Belgium
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
- Department of Internal Medicine (Cardiology), Yale School of Medicine, New Haven, CT 06511
- Department of Physiology, Yale School of Medicine, New Haven, CT 06511
| | - Fang Liu
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-8554
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-8554
- Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-8554
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-8554
| | - Bing Su
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
- Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Michael Simons
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06511
- Department of Internal Medicine (Cardiology), Yale School of Medicine, New Haven, CT 06511
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511;
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06511
- Department of Internal Medicine (Cardiology), Yale School of Medicine, New Haven, CT 06511
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520
| |
Collapse
|
39
|
Padmanaban P, Chizari A, Knop T, Zhang J, Trikalitis VD, Koopman B, Steenbergen W, Rouwkema J. Assessment of flow within developing chicken vasculature and biofabricated vascularized tissues using multimodal imaging techniques. Sci Rep 2021; 11:18251. [PMID: 34521868 PMCID: PMC8440514 DOI: 10.1038/s41598-021-97008-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/19/2021] [Indexed: 11/21/2022] Open
Abstract
Fluid flow shear stresses are strong regulators for directing the organization of vascular networks. Knowledge of structural and flow dynamics information within complex vasculature is essential for tuning the vascular organization within engineered tissues, by manipulating flows. However, reported investigations of vascular organization and their associated flow dynamics within complex vasculature over time are limited, due to limitations in the available physiological pre-clinical models, and the optical inaccessibility and aseptic nature of these models. Here, we developed laser speckle contrast imaging (LSCI) and side-stream dark field microscopy (SDF) systems to map the vascular organization, spatio-temporal blood flow fluctuations as well as erythrocytes movements within individual blood vessels of developing chick embryo, cultured within an artificial eggshell system. By combining imaging data and computational simulations, we estimated fluid flow shear stresses within multiscale vasculature of varying complexity. Furthermore, we demonstrated the LSCI compatibility with bioengineered perfusable muscle tissue constructs, fabricated via molding techniques. The presented application of LSCI and SDF on perfusable tissues enables us to study the flow perfusion effects in a non-invasive fashion. The gained knowledge can help to use fluid perfusion in order to tune and control multiscale vascular organization within engineered tissues.
Collapse
Affiliation(s)
- Prasanna Padmanaban
- Vascularization Lab, Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Ata Chizari
- Biomedical Photonic Imaging, Technical Medical Centre, Faculty of Science and Technology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Tom Knop
- Biomedical Photonic Imaging, Technical Medical Centre, Faculty of Science and Technology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Jiena Zhang
- Vascularization Lab, Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Vasileios D Trikalitis
- Vascularization Lab, Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Bart Koopman
- Vascularization Lab, Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Wiendelt Steenbergen
- Biomedical Photonic Imaging, Technical Medical Centre, Faculty of Science and Technology, University of Twente, 7500 AE, Enschede, The Netherlands.
| | - Jeroen Rouwkema
- Vascularization Lab, Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, 7500 AE, Enschede, The Netherlands.
| |
Collapse
|
40
|
James BD, Allen JB. Sex-Specific Response to Combinations of Shear Stress and Substrate Stiffness by Endothelial Cells In Vitro. Adv Healthc Mater 2021; 10:e2100735. [PMID: 34142471 PMCID: PMC8458248 DOI: 10.1002/adhm.202100735] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 12/25/2022]
Abstract
By using a full factorial design of experiment, the combinatorial effects of biological sex, shear stress, and substrate stiffness on human umbilical vein endothelial cell (HUVEC) spreading and Yes-associated protein 1 (YAP1) activity are able to be efficiently evaluated. Within the range of shear stress (0.5-1.5 Pa) and substrate stiffness (10-100 kPa), male HUVECs are smaller than female HUVECs. Only with sufficient mechanical stimulation do they spread to a similar size. More importantly, YAP1 nuclear localization in female HUVECs is invariant to mechanical stimulation within the range of tested conditions whereas for male HUVECs it increases nonlinearly with increasing shear stress and substrate stiffness. The sex-specific response of HUVECs to combinations of shear stress and substrate stiffness reinforces the need to include sex as a biological variable and multiple mechanical stimuli in experiments, informs the design of precision biomaterials, and offers insight for understanding cardiovascular disease sexual dimorphisms. Moreover, here it is illustrated that different complex mechanical microenvironments can lead to sex-specific phenotypes and sex invariant phenotypes in cultured endothelial cells.
Collapse
Affiliation(s)
- Bryan D James
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| |
Collapse
|
41
|
Dawson A, Wang Y, Li Y, LeMaire SA, Shen YH. New Technologies With Increased Precision Improve Understanding of Endothelial Cell Heterogeneity in Cardiovascular Health and Disease. Front Cell Dev Biol 2021; 9:679995. [PMID: 34513826 PMCID: PMC8430032 DOI: 10.3389/fcell.2021.679995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells (ECs) are vital for blood vessel integrity and have roles in maintaining normal vascular function, healing after injury, and vascular dysfunction. Extensive phenotypic heterogeneity has been observed among ECs of different types of blood vessels in the normal and diseased vascular wall. Although ECs with different phenotypes can share common functions, each has unique features that may dictate a fine-tuned role in vascular health and disease. Recent studies performed with single-cell technology have generated powerful information that has significantly improved our understanding of EC biology. Here, we summarize a variety of EC types, states, and phenotypes recently identified by using new, increasingly precise techniques in transcriptome analysis.
Collapse
Affiliation(s)
- Ashley Dawson
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
42
|
Stockinger B, Shah K, Wincent E. AHR in the intestinal microenvironment: safeguarding barrier function. Nat Rev Gastroenterol Hepatol 2021; 18:559-570. [PMID: 33742166 PMCID: PMC7611426 DOI: 10.1038/s41575-021-00430-8] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/01/2023]
Abstract
Mammalian aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that belongs to the basic helix-loop-helix (bHLH)-PAS family of transcription factors, which are evolutionarily conserved environmental sensors. In the absence of ligands, AHR resides in the cytoplasm in a complex with molecular chaperones such as HSP90, XAP2 and p23. Upon ligand binding, AHR translocates into the nuclear compartment, where it dimerizes with its partner protein, AHR nuclear translocator (ARNT), an obligatory partner for the DNA-binding and functional activity. Historically, AHR had mostly been considered as a key intermediary for the detrimental effects of environmental pollutants on the body. However, following the discovery of AHR-mediated functions in various immune cells, as well as the emergence of non-toxic 'natural' AHR ligands, this view slowly began to change, and the study of AHR-deficient mice revealed a plethora of important beneficial functions linked to AHR activation. This Review focuses on regulation of the AHR pathway and the barrier-protective roles AHR has in haematopoietic, as well as non-haematopoietic, cells within the intestinal microenvironment. It covers the nature of AHR ligands and feedback regulation of the AHR pathway, outlining the currently known physiological functions in immune, epithelial, endothelial and neuronal cells of the intestine.
Collapse
Affiliation(s)
| | | | - Emma Wincent
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
43
|
Abstract
The lymphatic vasculature plays important role in regulating fluid homeostasis, intestinal lipid absorption, and immune surveillance in humans. Malfunction of lymphatic vasculature leads to several human diseases. Understanding the fundamental mechanism in lymphatic vascular development not only expand our knowledge, but also provide a new therapeutic insight. Recently, Hippo-YAP/TAZ signaling pathway, a key mechanism of organ size and tissue homeostasis, has emerged as a critical player that regulate lymphatic specification, sprouting, and maturation. In this review, we discuss the mechanistic regulation and pathophysiological significant of Hippo pathway in lymphatic vascular development.
Collapse
Affiliation(s)
- Boksik Cha
- Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Sungjin Moon
- Department of Biological Science, Kangwon National University, Chuncheon 24341, Korea
| | - Wantae Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
44
|
Dysregulated Expression of Arterial MicroRNAs and Their Target Gene Networks in Temporal Arteries of Treatment-Naïve Patients with Giant Cell Arteritis. Int J Mol Sci 2021; 22:ijms22126520. [PMID: 34204585 PMCID: PMC8234166 DOI: 10.3390/ijms22126520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
In this study, we explored expression of microRNA (miR), miR-target genes and matrix remodelling molecules in temporal artery biopsies (TABs) from treatment-naïve patients with giant cell arteritis (GCA, n = 41) and integrated these analyses with clinical, laboratory, ultrasound and histological manifestations of GCA. NonGCA patients (n = 4) served as controls. GCA TABs exhibited deregulated expression of several miRs (miR-21-5p, -145-5p, -146a-5p, -146b-5p, -155-5p, 424-3p, -424-5p, -503-5p), putative miR-target genes (YAP1, PELI1, FGF2, VEGFA, KLF4) and matrix remodelling factors (MMP2, MMP9, TIMP1, TIPM2) with key roles in Toll-like receptor signaling, mechanotransduction and extracellular matrix biology. MiR-424-3p, -503-5p, KLF4, PELI1 and YAP1 were identified as new deregulated molecular factors in GCA TABs. Quantities of miR-146a-5p, YAP1, PELI1, FGF2, TIMP2 and MMP9 were particularly high in histologically positive GCA TABs with occluded temporal artery lumen. MiR-424-5p expression in TABs and the presence of facial or carotid arteritis on ultrasound were associated with vision disturbances in GCA patients. Correlative analysis of miR-mRNA quantities demonstrated a highly interrelated expression network of deregulated miRs and mRNAs in temporal arteries and identified KLF4 as a candidate target gene of deregulated miR-21-5p, -146a-5p and -155-5p network in GCA TABs. Meanwhile, arterial miR and mRNA expression did not correlate with constitutive symptoms and signs of GCA, elevated markers of systemic inflammation nor sonographic characteristics of GCA. Our study provides new insights into GCA pathophysiology and uncovers new candidate biomarkers of vision impairment in GCA.
Collapse
|
45
|
Liu Q, Zhang Y, Yang J, Yang Y, Li M, Chen S, Jiang P, Wang N, Zhang Y, Liu J, Wu J, Wang S. The Relationship of Morphological-Hemodynamic Characteristics, Inflammation, and Remodeling of Aneurysm Wall in Unruptured Intracranial Aneurysms. Transl Stroke Res 2021; 13:88-99. [PMID: 34105079 DOI: 10.1007/s12975-021-00917-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Aneurysm wall remodeling (AWR) is an important pathological characteristic in aneurysm wall, which was characterized by abnormal histological structure and inflammation infiltration. In the present study, the aim is to determine the relationships of morphological-hemodynamic characteristics, inflammation, and AWR in intracranial aneurysms (IAs), as well as the pathological basis of morphological-hemodynamic predictors to achieve IA development. For this end, 113 unruptured IAs were prospectively collected from 110 cases. In addition, patient-specific computational fluid dynamics and geometry were adopted to determine hemodynamic and morphological parameters. Moreover, Hematoxylin-Eosin staining was performed to identify the AWR. By performing immunofluorescence, the inflammatory markers were detected. Masson staining was conducted to characterize the characteristics of atherosclerosis in aneurysm wall. To demonstrate the parameters regarding the AWR, a multivariate logistic analysis was conducted. Besides, correlation analyses were conducted to verify the relationship between morphological-hemodynamic and pathological characteristics. For 113 unruptured IAs, no difference was identified in baseline information. AWR was demonstrated in 92 (81.4%) IAs. To be specific, the aneurysm size (odds ratio (OR), 2.63; confidence interval (CI), 1.04-6.67; P = 0.041), size ratio (SR; OR, 1.95; CI, 1.38-2.76; P < 0.001), normalized wall shear stress average (NWSSA; OR, 0.05; CI, 0.01-0.15; P = 0.007), and relative resident time (RRT; OR, 1.28; CI, 1.07-1.53; P = 0.007) were proved as the factors of AWR. As revealed from the results of immunofluorescence, aneurysm size, SR, NWSSA, and RRT were significantly correlated with the level of inflammation in IA tissues. Furthermore, Masson staining revealed that atherosclerosis area in IA tissues and NWSSA was correlated with RRT. In this study, SR, NWSSA, and RRT were demonstrated as the risk factors of AWR. The mentioned parameters could also reflect the characteristics of inflammation and atherosclerosis in aneurysm wall as well. This study revealed that biomechanical stress and inflammation in aneurysm wall are correlated, which might suggest the pathological evidence of morphological-hemodynamic predictors for IA development.
Collapse
Affiliation(s)
- Qingyuan Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yisen Zhang
- Department of Neurointervention, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
| | - Junhua Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yi Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Maogui Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shanwen Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Pengjun Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Nuochuan Wang
- Department of Blood Transfusion, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
| | - Yanan Zhang
- Department of Blood Transfusion, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
| | - Jia Liu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 85411, Guangdong, China
| | - Jun Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
46
|
Shear forces induce ICAM-1 nanoclustering on endothelial cells that impact on T-cell migration. Biophys J 2021; 120:2644-2656. [PMID: 34087211 DOI: 10.1016/j.bpj.2021.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 01/13/2023] Open
Abstract
The leukocyte-specific β2-integrin LFA-1 and its ligand ICAM-1, expressed on endothelial cells (ECs), are involved in the arrest, adhesion, and transendothelial migration of leukocytes. Although the role of mechanical forces on LFA-1 activation is well established, the impact of forces on its major ligand ICAM-1 has received less attention. Using a parallel-plate flow chamber combined with confocal and super-resolution microscopy, we show that prolonged shear flow induces global translocation of ICAM-1 on ECs upstream of flow direction. Interestingly, shear forces caused actin rearrangements and promoted actin-dependent ICAM-1 nanoclustering before LFA-1 engagement. T cells adhered to mechanically prestimulated ECs or nanoclustered ICAM-1 substrates developed a promigratory phenotype, migrated faster, and exhibited shorter-lived interactions with ECs than when adhered to non mechanically stimulated ECs or to monomeric ICAM-1 substrates. Together, our results indicate that shear forces increase ICAM-1/LFA-1 bonds because of ICAM-1 nanoclustering, strengthening adhesion and allowing cells to exert higher traction forces required for faster migration. Our data also underscore the importance of mechanical forces regulating the nanoscale organization of membrane receptors and their contribution to cell adhesion regulation.
Collapse
|
47
|
Schofield CL, Rodrigo-Navarro A, Dalby MJ, Van Agtmael T, Salmeron-Sanchez M. Biochemical‐ and Biophysical‐Induced Barriergenesis in the Blood–Brain Barrier: A Review of Barriergenic Factors for Use in In Vitro Models. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
| | | | - Matthew J. Dalby
- Centre for the Cellular Microenvironment University of Glasgow Glasgow UK
| | - Tom Van Agtmael
- Institute of Cardiovascular and Medical Sciences University of Glasgow Glasgow UK
| | | |
Collapse
|
48
|
Liu M, Wang D, Gu S, Tian B, Liang J, Suo Q, Zhang Z, Yang G, Zhou Y, Li S. Micro/nano materials regulate cell morphology and intercellular communication by extracellular vesicles. Acta Biomater 2021; 124:130-138. [PMID: 33567350 DOI: 10.1016/j.actbio.2021.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) have emerged as important nano-cargo carriers for cell-cell communication, yet how biophysical factors regulate EV-mediated signaling is not well understood. Here we show that microgrooves can modulate the morphology of endothelial cells (ECs), and regulate the phenotype of smooth muscle cells (SMCs) through EVs in co-culture. Elongated ECs, in comparison with polygonal ECs, increased the expression of contractile markers in SMCs. Depletion of EVs in the culture medium abolished this effect. Further analysis demonstrated that elongated ECs significantly upregulated miR-143/miR-145, leading to the increase of these microRNAs in EC-secreted EVs that were transferred to SMCs under a co-culture condition. Inhibition of EV secretion from ECs abolished the EC-SMC communication and the increased expression of SMC contractile markers. Moreover, electrospun nano-fibrous scaffolds with aligned fibers had the same effects as microgrooves to induce EC secretion of EVs to regulate SMC phenotypic marker expression. These results demonstrate that micro and nano materials can be used to engineer cell morphology and regulate EV secretion for cell-cell communication, which will have significant implications in the engineering of blood vessels and other tissues. STATEMENT OF SIGNIFICANCE: By manipulating EC morphology with micro/nano materials, we show that EV-mediated signaling can regulate SMC phenotypic marker expression. This is a very thorough and unique study to demonstrate the function of extracellular vesicles (EVs) as important nano-carriers in cell-cell communication. The originality of this study is to demonstrate that EC morphology modulates the phenotype of smooth muscle cells via extracellular vesicles enclosing miR143/miR145. These findings underscore the important role of biophysical changes in cell-cell communications, and provide a rational basis for engineering micro/nano materials to control cell-cell communications for cell and tissue engineering.
Collapse
Affiliation(s)
- Mengya Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Dan Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Shuangying Gu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Baoxiang Tian
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Jiaqi Liang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Qian Suo
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Zhijun Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Guoyuan Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Yue Zhou
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | - Song Li
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, CA 90095, United States
| |
Collapse
|
49
|
Abutaleb NO, Truskey GA. Differentiation and characterization of human iPSC-derived vascular endothelial cells under physiological shear stress. STAR Protoc 2021; 2:100394. [PMID: 33796871 PMCID: PMC7995664 DOI: 10.1016/j.xpro.2021.100394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer a potentially unlimited source to generate endothelial cells (ECs) for numerous applications. Here, we describe a 7-day protocol to differentiate up to 55 million vascular endothelial cells (viECs) from 3.5 million human iPSCs using small molecules to regulate specific transcription factors. We also describe a parallel-plate flow chamber system to study EC behavior under physiological shear stress. For complete details on the use and execution of this protocol, please refer to Atchison et al. (2020). A protocol for differentiating vascular endothelial cells (viECs) from human iPSCs Generation of up to 55 million viECs from 3.5 million iPSCs within 7 days Design and use of parallel-plate flow chamber to study EC behavior under flow viECs express EC markers, upregulate flow-sensitive genes, and align to flow direction
Collapse
Affiliation(s)
- Nadia O Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC 27713, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27713, USA
| |
Collapse
|
50
|
Tanaka K, Joshi D, Timalsina S, Schwartz MA. Early events in endothelial flow sensing. Cytoskeleton (Hoboken) 2021; 78:217-231. [PMID: 33543538 DOI: 10.1002/cm.21652] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Responses of vascular and lymphatic endothelial cells (ECs) to fluid shear stress (FSS) from blood or lymphatic fluid flow govern the development, physiology, and diseases of these structures. Extensive research has characterized the signaling, gene expression and cytoskeletal pathways that mediate effects on EC phenotype and vascular morphogenesis. But the primary mechanisms by which ECs transduce the weak forces from flow into biochemical signals are less well understood. This review covers recent advances in our understanding of the immediate mechanisms of FSS mechanotransduction, integrating results from different disciplines, addressing their roles in development, physiology and disease, and suggesting important questions for future work.
Collapse
Affiliation(s)
- Keiichiro Tanaka
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Divyesh Joshi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Sushma Timalsina
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA.,Department of Cell Biology, Yale University, New Haven, Connecticut, USA.,Department of Biomedical engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|