1
|
Hou HT, Chen HX, Wang ZQ, Xi L, Wang J, Yang Q, He GW. Arterial and venous grafting biomaterials in coronary Surgery: Integrative multi-omics approach reveals ECM-PI3K-Akt pathway as Key Regulator of different patency. CHEMICAL ENGINEERING JOURNAL 2025; 511:161829. [DOI: 10.1016/j.cej.2025.161829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
|
2
|
Sun D, Zhang K, Zheng F, Yang G, Yang M, Xu Y, Qin Y, Lin M, Li Y, Tan J, Li Q, Qu X, Li G, Bian L, Zhu C. Matrix Viscoelasticity Controls Differentiation of Human Blood Vessel Organoids into Arterioles and Promotes Neovascularization in Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2410802. [PMID: 39686788 DOI: 10.1002/adma.202410802] [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: 07/24/2024] [Revised: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Stem cell-derived blood vessel organoids are embedded in extracellular matrices to stimulate vessel sprouting. Although vascular organoids in 3D collagen I-Matrigel gels are currently available, they are primarily capillaries composed of endothelial cells (ECs), pericytes, and mesenchymal stem-like cells, which necessitate mature arteriole differentiation for neovascularization. In this context, the hypothesis that matrix viscoelasticity regulates vascular development is investigated in 3D cultures by encapsulating blood vessel organoids within viscoelastic gelatin/β-CD assembly dynamic hydrogels or methacryloyl gelatin non-dynamic hydrogels. The vascular organoids within the dynamic hydrogel demonstrate enhanced angiogenesis and differentiation into arterioles containing smooth muscle cells. The dynamic hydrogel mechanical microenvironment promotes vascular patterning and arteriolar differentiation by elevating notch receptor 3 signaling in mesenchymal stem cells and downregulating platelet-derived growth factor B expression in ECs. Transplantation of vascular organoids in vivo, along with the dynamic hydrogel, leads to the reassembly of arterioles and restoration of cardiac function in infarcted hearts. These findings indicate that the viscoelastic properties of the matrix play a crucial role in controlling the vascular organization and differentiation processes, suggesting an exciting potential for its application in regenerative medicine.
Collapse
Affiliation(s)
- Dayu Sun
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Feiyang Zheng
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Guanyuan Yang
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Mingcan Yang
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Youqian Xu
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Yinhua Qin
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Mingxin Lin
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yanzhao Li
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Ju Tan
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Qiyu Li
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Xiaohang Qu
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Gang Li
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chuhong Zhu
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| |
Collapse
|
3
|
Xing M, Wang F, Chu R, Wang H, Sun Y, Qian M, Jiang H, Midgley AC, Dai G, Zhao Q. Localized COUP-TFII pDNA Delivery Modulates Stem/Progenitor Cell Differentiation to Enhance Endothelialization and Inhibit Calcification of Decellularized Allografts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409744. [PMID: 39656938 PMCID: PMC11792037 DOI: 10.1002/advs.202409744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/12/2024] [Indexed: 12/17/2024]
Abstract
Decellularized allografts have emerged as promising candidates for vascular bypass grafting, owing to their inherent bioactivity and minimal immunogenicity. However, graft failure that results from suboptimal regeneration and pathological remodeling has hindered their clinical adoption. Recent advances in vascular biology highlight the pivotal role of COUP-TFII in orchestrating endothelial identity, angiogenesis, safeguarding against atherosclerosis, and mitigating vascular calcification. Here, plasmid DNA (pDNA) encoding COUP-TFII is incorporated into decellularized allografts to realize localized delivery. Comprehensive in vitro investigation complemented by a bone marrow transplantation model on genetic-lineage-tracing mouse revealed the underlying mechanisms of COUP-TFII in regulating vascular regeneration and remodeling. COUP-TFII augmented endothelialization and inhibited calcification in decellularized allografts by modulating the Ang1/Tie2/PI3K/AKT signaling pathway that dictates the fate of Sca-1+ stem/progenitor cells. Heparin-polyethyleneimine nanoparticles (HEPI) are prepared as COUP-TFII pDNA nanocarriers (COUP-TFII@HPEI) and used to modify decellularized allografts, achieving long-term and stable overexpression of COUP-TFII. Functionalized grafts are evaluated in rat abdominal artery replacement models, demonstrating enhanced neo-artery regeneration without calcification. The study provides an effective strategy to enhance the applicability of decellularized allograft and illustrates their translational prospects for vascular bypass grafting.
Collapse
Affiliation(s)
- Mengmeng Xing
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Fei Wang
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Ruowen Chu
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - He Wang
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Yuyao Sun
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Meng Qian
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Huan Jiang
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Guohao Dai
- Department of BioengineeringNortheastern UniversityBostonMA02115USA
| | - Qiang Zhao
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and RemodelingHealth Science CenterPeking UniversityBeijing100191China
| |
Collapse
|
4
|
Luo X, Pang Z, Li J, Anh M, Kim BS, Gao G. Bioengineered human arterial equivalent and its applications from vascular graft to in vitro disease modeling. iScience 2024; 27:111215. [PMID: 39555400 PMCID: PMC11565542 DOI: 10.1016/j.isci.2024.111215] [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] [Indexed: 11/19/2024] Open
Abstract
Arterial disorders such as atherosclerosis, thrombosis, and aneurysm pose significant health risks, necessitating advanced interventions. Despite progress in artificial blood vessels and animal models aimed at understanding pathogenesis and developing therapies, limitations in graft functionality and species discrepancies restrict their clinical and research utility. Addressing these issues, bioengineered arterial equivalents (AEs) with enhanced vascular functions have been developed, incorporating innovative technologies that improve clinical outcomes and enhance disease progression modeling. This review offers a comprehensive overview of recent advancements in bioengineered AEs, systematically summarizing the bioengineered technologies used to construct these AEs, and discussing their implications for clinical application and pathogenesis understanding. Highlighting current breakthroughs and future perspectives, this review aims to inform and inspire ongoing research in the field, potentially transforming vascular medicine and offering new avenues for preclinical and clinical advances.
Collapse
Affiliation(s)
- Xi Luo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zherui Pang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology, Zhuhai 519088, China
| | - Minjun Anh
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
5
|
Szukiewicz D. CX3CL1 (Fractalkine)-CX3CR1 Axis in Inflammation-Induced Angiogenesis and Tumorigenesis. Int J Mol Sci 2024; 25:4679. [PMID: 38731899 PMCID: PMC11083509 DOI: 10.3390/ijms25094679] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The chemotactic cytokine fractalkine (FKN, chemokine CX3CL1) has unique properties resulting from the combination of chemoattractants and adhesion molecules. The soluble form (sFKN) has chemotactic properties and strongly attracts T cells and monocytes. The membrane-bound form (mFKN) facilitates diapedesis and is responsible for cell-to-cell adhesion, especially by promoting the strong adhesion of leukocytes (monocytes) to activated endothelial cells with the subsequent formation of an extracellular matrix and angiogenesis. FKN signaling occurs via CX3CR1, which is the only known member of the CX3C chemokine receptor subfamily. Signaling within the FKN-CX3CR1 axis plays an important role in many processes related to inflammation and the immune response, which often occur simultaneously and overlap. FKN is strongly upregulated by hypoxia and/or inflammation-induced inflammatory cytokine release, and it may act locally as a key angiogenic factor in the highly hypoxic tumor microenvironment. The importance of the FKN/CX3CR1 signaling pathway in tumorigenesis and cancer metastasis results from its influence on cell adhesion, apoptosis, and cell migration. This review presents the role of the FKN signaling pathway in the context of angiogenesis in inflammation and cancer. The mechanisms determining the pro- or anti-tumor effects are presented, which are the cause of the seemingly contradictory results that create confusion regarding the therapeutic goals.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
6
|
Loh KM, Ang LT. Building human artery and vein endothelial cells from pluripotent stem cells, and enduring mysteries surrounding arteriovenous development. Semin Cell Dev Biol 2024; 155:62-75. [PMID: 37393122 DOI: 10.1016/j.semcdb.2023.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Owing to their manifold roles in health and disease, there have been intense efforts to synthetically generate blood vessels in vitro from human pluripotent stem cells (hPSCs). However, there are multiple types of blood vessel, including arteries and veins, which are molecularly and functionally different. How can we specifically generate either arterial or venous endothelial cells (ECs) from hPSCs in vitro? Here, we summarize how arterial or venous ECs arise during embryonic development. VEGF and NOTCH arbitrate the bifurcation of arterial vs. venous ECs in vivo. While manipulating these two signaling pathways biases hPSC differentiation towards arterial and venous identities, efficiently generating these two subtypes of ECs has remained challenging until recently. Numerous questions remain to be fully addressed. What is the complete identity, timing and combination of extracellular signals that specify arterial vs. venous identities? How do these extracellular signals intersect with fluid flow to modulate arteriovenous fate? What is a unified definition for endothelial progenitors or angioblasts, and when do arterial vs. venous potentials segregate? How can we regulate hPSC-derived arterial and venous ECs in vitro, and generate organ-specific ECs? In turn, answers to these questions could avail the production of arterial and venous ECs from hPSCs, accelerating vascular research, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Kyle M Loh
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA.
| | - Lay Teng Ang
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Yang X, Sun S, Zhou Z, Chen X, Chen G. Continuous Extrusion Forming Technology of Magnesium Alloy Thin-Walled Tubules. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5803. [PMID: 37687500 PMCID: PMC10488630 DOI: 10.3390/ma16175803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023]
Abstract
This paper proposes a new technology of superimposed billet extrusion-forming for thin-walled magnesium alloy tubes. This process represents an improvement over the current technology, which suffers from low production efficiency, poor forming accuracy, and low material utilization. We developed a detailed forming process and mold structure, in which the excess material of the front billet is extruded out of the mold as the rear billet pushes on the front one. Through continuous extrusion, online direct water cooling, and cutting, the automated continuous production of thin-walled tubules is achieved. The optimization of the mandrel structure and its hovering action is also included, with the aim of improving the lifespan of the mandrel and the accuracy of tube size. The numerical simulation method evaluates the effect of the die angle (α) on the tube, formed using FORGE NXT 1.1. The results show that for an angle of less than 70°, the defect length of the tube decreases as the die angle decreases, forming an ordered flow of superimposed billets. If the angle is less than 50°, the two adjacently formed tubes separate automatically, with no need for the subsequent cutting process. The best choice of die angle is about 50°, which takes into account the effect of the change in extrusion force.
Collapse
Affiliation(s)
- Xi Yang
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, China
- Collaborative Innovation Center of Nonferrous Metals of Henan Province, Luoyang 471023, China
| | - Shihan Sun
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, China
- Collaborative Innovation Center of Nonferrous Metals of Henan Province, Luoyang 471023, China
| | - Zheng Zhou
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, China
- Collaborative Innovation Center of Nonferrous Metals of Henan Province, Luoyang 471023, China
| | - Xuewen Chen
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, China
- Collaborative Innovation Center of Nonferrous Metals of Henan Province, Luoyang 471023, China
| | - Guoqing Chen
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China;
| |
Collapse
|
8
|
Liu Z, Huang Y, Wang D, Li M, Zhang Q, Pan C, Lin Y, Luo Y, Shi Z, Zhang P, Zheng Y. Insights gained from single-cell RNA analysis of murine endothelial cells in aging hearts. Heliyon 2023; 9:e18324. [PMID: 37554834 PMCID: PMC10404962 DOI: 10.1016/j.heliyon.2023.e18324] [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: 01/24/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/10/2023] Open
Abstract
Aging is the strongest risk factor for cardiovascular disease, with progressive decline in the function of vascular endothelial cells (ECs) with age. Systematic analyses of the effects of aging on different cardiac EC types remain limited. Here, we constructed a scRNA atlas of EC transcriptomes in young and old mouse hearts. We identified 10 EC subclusters. The multidimensionally differential genes (DEGs) analysis across different EC clusters shows molecular changes with aging, showing the increase in the overall inflammatory microenvironment and the decrease in angiogenesis and cytoskeletal support capacity of aged ECs. And we performed an in-depth analysis of 3 special ECs, Immunology, Proliferating and Angiogenic. The Immunology EC seems highly associated with some immune regulatory functions, which decline with aging at different degrees. Analysis of two types of neovascular ECs, Proliferating, Angiogenic, implied that Angiogenic ECs can differentiate into multiple EC directions after initially originating from proliferating ECs. And aging leads to a decrease in the ability of vascular angiogenesis and differentiation. Finally, we summarized the effects of aging on cell signaling communication between different EC clusters. This cardiac EC atlas offers comprehensive insights into the molecular regulations of cardiovascular aging, and provides new directions for the prevention and treatment of age-related cardiovascular disease.
Collapse
Affiliation(s)
- Zhong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100085, China
| | - Yanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dongliang Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mengke Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Qikai Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Caineng Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yuheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yuanting Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Zhuoxing Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yingfeng Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100085, China
| |
Collapse
|
9
|
Laboyrie SL, de Vries MR, Bijkerk R, Rotmans JI. Building a Scaffold for Arteriovenous Fistula Maturation: Unravelling the Role of the Extracellular Matrix. Int J Mol Sci 2023; 24:10825. [PMID: 37446003 DOI: 10.3390/ijms241310825] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Vascular access is the lifeline for patients receiving haemodialysis as kidney replacement therapy. As a surgically created arteriovenous fistula (AVF) provides a high-flow conduit suitable for cannulation, it remains the vascular access of choice. In order to use an AVF successfully, the luminal diameter and the vessel wall of the venous outflow tract have to increase. This process is referred to as AVF maturation. AVF non-maturation is an important limitation of AVFs that contributes to their poor primary patency rates. To date, there is no clear overview of the overall role of the extracellular matrix (ECM) in AVF maturation. The ECM is essential for vascular functioning, as it provides structural and mechanical strength and communicates with vascular cells to regulate their differentiation and proliferation. Thus, the ECM is involved in multiple processes that regulate AVF maturation, and it is essential to study its anatomy and vascular response to AVF surgery to define therapeutic targets to improve AVF maturation. In this review, we discuss the composition of both the arterial and venous ECM and its incorporation in the three vessel layers: the tunica intima, media, and adventitia. Furthermore, we examine the effect of chronic kidney failure on the vasculature, the timing of ECM remodelling post-AVF surgery, and current ECM interventions to improve AVF maturation. Lastly, the suitability of ECM interventions as a therapeutic target for AVF maturation will be discussed.
Collapse
Affiliation(s)
- Suzanne L Laboyrie
- Department of Internal Medicine, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Vascular Surgery, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
10
|
Ma Y, Zhu Q, Luo S, Zhang F, Liu L, Mengxue Z, Zhang Z, Cao X, Qiu X, Zeng X, Ji D, Li C, Zhong X, Wang J, Wei Y. Environmentally relevant concentrations of fipronil selectively disrupt venous vessel development in zebrafish embryos/larvae. CHEMOSPHERE 2023:139146. [PMID: 37290517 DOI: 10.1016/j.chemosphere.2023.139146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/29/2023] [Accepted: 06/04/2023] [Indexed: 06/10/2023]
Abstract
The pesticide fipronil is widely dispersed in aquatic environments and frequently detected in the general population. Although the adverse effects on embryonic growth by fipronil exposure have been extensively documented, the early responses for its developmental toxicity are largely unknown. In the present study, we explored the sensitive targets of fipronil, focusing on vascular injury using zebrafish embryos/larvae and cultured human endothelial cells. Exposure to 5-500 μg/L fipronil at the early stage impeded the growth of sub-intestinal venous plexus (SIVP), caudal vein plexus (CVP), and common cardinal veins (CCV). The damages on venous vessels occurred at exposure to the environmentally relevant concentration as low as 5 μg/L fipronil, whereas no significant change was observed in general toxicity indexes. In contrast, vascular development of the dorsal aorta (DA) or intersegmental artery (ISA) was not affected. In addition, the mRNA levels of vascular markers and vessel type-specific function genes exhibited significant decreases in venous genes, including nr2f2, ephb4a, and flt4, but no appreciable change in arterial genes. Likewise, the more pronounced changes in cell death and cytoskeleton disruption were shown in human umbilical vein endothelial cells as compared with human aortic endothelial cells. Furthermore, molecular docking supported a stronger affinity of fipronil and its metabolites to the proteins correlated with venous development, such as BMPR2 and SMARCA4. These results reveal the heterogeneity in developing vasculature responsive to fipronil's exposure. The preferential impacts on the veins confer higher sensitivity, allowing them to be appropriate targets for monitoring fipronil's developmental toxicity.
Collapse
Affiliation(s)
- Ya Ma
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qicheng Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shili Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fenghong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Lei Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Zhi Mengxue
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Zhuyi Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaolian Cao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xuelin Qiu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiangyu Zeng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Di Ji
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chenxin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiali Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jianshe Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| | - Yanhong Wei
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
11
|
Zhang R, Liu Q, Lyu C, Gao X, Ma W. Knockdown SENP1 Suppressed the Angiogenic Potential of Mesenchymal Stem Cells by Impacting CXCR4-Regulated MRTF-A SUMOylation and CCN1 Expression. Biomedicines 2023; 11:biomedicines11030914. [PMID: 36979893 PMCID: PMC10046070 DOI: 10.3390/biomedicines11030914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
The angiogenic potential of mesenchymal stem cells (MSCs) is critical for adult vascular regeneration and repair, which is regulated by various growth factors and cytokines. In the current study, we report that knockdown SUMO-specific peptidase 1 (SENP1) stimulated the SUMOylation of MRTF-A and prevented its translocation into the nucleus, leading to downregulation of the cytokine and angiogenic factor CCN1, which significantly impacted MSC-mediated angiogenesis and cell migration. Further studies showed that SENP1 knockdown also suppressed the expression of a chemokine receptor CXCR4, and overexpression of CXCR4 could partially abrogate MRTF-A SUMOylation and reestablish the CCN1 level. Mutation analysis confirmed that SUMOylation occurred on three lysine residues (Lys-499, Lys-576, and Lys-624) of MRTF-A. In addition, SENP1 knockdown abolished the synergistic co-activation of CCN1 between MRTF-A and histone acetyltransferase p300 by suppressing acetylation on histone3K9, histone3K14, and histone4. These results revealed an important signaling pathway to regulate MSC differentiation and angiogenesis by MRTF-A SUMOylation involving cytokine/chemokine activities mediated by CCN1 and CXCR4, which may potentially impact a variety of cellular processes such as revascularization, wound healing, and progression of cancer.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Hematology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Qingxi Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
- Department of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
- Correspondence: (Q.L.); (W.M.)
| | - Cuicui Lyu
- Department of Hematology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xing Gao
- Department of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
| | - Wenjian Ma
- Department of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Correspondence: (Q.L.); (W.M.)
| |
Collapse
|
12
|
Lin HH, Kuo MW, Fan TC, Yu AL, Yu J. YULINK regulates vascular formation in zebrafish and HUVECs. Biol Res 2023; 56:7. [PMID: 36843032 PMCID: PMC9969694 DOI: 10.1186/s40659-023-00415-8] [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: 08/24/2022] [Accepted: 01/18/2023] [Indexed: 02/28/2023] Open
Abstract
BACKGROUND The distinct arterial and venous cell fates are dictated by a combination of various genetic factors which form diverse types of blood vessels such as arteries, veins, and capillaries. We report here that YULINK protein is involved in vasculogenesis, especially venous formation. METHODS In this manuscript, we employed gene knockdown, yeast two-hybrid, FLIM-FRET, immunoprecipitation, and various imaging technologies to investigate the role of YULINK gene in zebrafish and human umbilical vein endothelial cells (HUVECs). RESULTS Knockdown of YULINK during the arterial-venous developmental stage of zebrafish embryos led to the defective venous formation and abnormal vascular plexus formation. Knockdown of YULINK in HUVECs impaired their ability to undergo cell migration and differentiation into a capillary-like tube formation. In addition, the phosphorylated EPHB4 was decreased in YULINK knockdown HUVECs. Yeast two-hybrid, FLIM-FRET, immunoprecipitation, as well as imaging technologies showed that YULINK colocalized with endosome related proteins (EPS15, RAB33B or TICAM2) and markers (Clathrin and RHOB). VEGF-induced VEGFR2 internalization was also compromised in YULINK knockdown HUVECs, demonstrating to the involvement of YULINK. CONCLUSION This study suggests that YULINK regulates vasculogenesis, possibly through endocytosis in zebrafish and HUVECs.
Collapse
Affiliation(s)
- Hsin-Hung Lin
- grid.28665.3f0000 0001 2287 1366Chemical Biology and Molecular Biophysics Program, International Graduate Program, Academia Sinica, Taipei, Taiwan ,grid.454210.60000 0004 1756 1461Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, 333 Taoyuan, Taiwan
| | - Ming-Wei Kuo
- grid.454210.60000 0004 1756 1461Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, 333 Taoyuan, Taiwan
| | - Tan-Chi Fan
- grid.454210.60000 0004 1756 1461Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, 333 Taoyuan, Taiwan
| | - Alice L. Yu
- grid.454210.60000 0004 1756 1461Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, 333 Taoyuan, Taiwan ,grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California, San Diego, CA USA
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, 333, Taoyuan, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
13
|
McCloskey MC, Zhang VZ, Ahmad SD, Walker S, Romanick SS, Awad HA, McGrath JL. Sourcing cells for in vitro models of human vascular barriers of inflammation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:979768. [PMID: 36483299 PMCID: PMC9724237 DOI: 10.3389/fmedt.2022.979768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 07/20/2023] Open
Abstract
The vascular system plays a critical role in the progression and resolution of inflammation. The contributions of the vascular endothelium to these processes, however, vary with tissue and disease state. Recently, tissue chip models have emerged as promising tools to understand human disease and for the development of personalized medicine approaches. Inclusion of a vascular component within these platforms is critical for properly evaluating most diseases, but many models to date use "generic" endothelial cells, which can preclude the identification of biomedically meaningful pathways and mechanisms. As the knowledge of vascular heterogeneity and immune cell trafficking throughout the body advances, tissue chip models should also advance to incorporate tissue-specific cells where possible. Here, we discuss the known heterogeneity of leukocyte trafficking in vascular beds of some commonly modeled tissues. We comment on the availability of different tissue-specific cell sources for endothelial cells and pericytes, with a focus on stem cell sources for the full realization of personalized medicine. We discuss sources available for the immune cells needed to model inflammatory processes and the findings of tissue chip models that have used the cells to studying transmigration.
Collapse
Affiliation(s)
- Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samantha S. Romanick
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
14
|
Liu C, Niu K, Xiao Q. Updated perspectives on vascular cell specification and pluripotent stem cell-derived vascular organoids for studying vasculopathies. Cardiovasc Res 2022; 118:97-114. [PMID: 33135070 PMCID: PMC8752356 DOI: 10.1093/cvr/cvaa313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Vasculopathy is a pathological process occurring in the blood vessel wall, which could affect the haemostasis and physiological functions of all the vital tissues/organs and is one of the main underlying causes for a variety of human diseases including cardiovascular diseases. Current pharmacological interventions aiming to either delay or stop progression of vasculopathies are suboptimal, thus searching novel, targeted, risk-reducing therapeutic agents, or vascular grafts with full regenerative potential for patients with vascular abnormalities are urgently needed. Since first reported, pluripotent stem cells (PSCs), particularly human-induced PSCs, have open new avenue in all research disciplines including cardiovascular regenerative medicine and disease remodelling. Assisting with recent technological breakthroughs in tissue engineering, in vitro construction of tissue organoid made a tremendous stride in the past decade. In this review, we provide an update of the main signal pathways involved in vascular cell differentiation from human PSCs and an extensive overview of PSC-derived tissue organoids, highlighting the most recent discoveries in the field of blood vessel organoids as well as vascularization of other complex tissue organoids, with the aim of discussing the key cellular and molecular players in generating vascular organoids.
Collapse
MESH Headings
- Blood Vessels/metabolism
- Blood Vessels/pathology
- Blood Vessels/physiopathology
- Cell Culture Techniques
- Cell Differentiation
- Cell Lineage
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Humans
- Induced Pluripotent Stem Cells/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Organoids
- Phenotype
- Signal Transduction
- Vascular Diseases/metabolism
- Vascular Diseases/pathology
- Vascular Diseases/physiopathology
Collapse
Affiliation(s)
- Chenxin Liu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Kaiyuan Niu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases at The Second Affiliated Hospital
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou, Guangdong 511436, China
| |
Collapse
|
15
|
Inverso D, Shi J, Lee KH, Jakab M, Ben-Moshe S, Kulkarni SR, Schneider M, Wang G, Komeili M, Vélez PA, Riedel M, Spegg C, Ruppert T, Schaeffer-Reiss C, Helm D, Singh I, Boutros M, Chintharlapalli S, Heikenwalder M, Itzkovitz S, Augustin HG. A spatial vascular transcriptomic, proteomic, and phosphoproteomic atlas unveils an angiocrine Tie-Wnt signaling axis in the liver. Dev Cell 2021; 56:1677-1693.e10. [PMID: 34038707 PMCID: PMC8191494 DOI: 10.1016/j.devcel.2021.05.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/18/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
Single-cell transcriptomics (scRNA-seq) has revolutionized the understanding of the spatial architecture of tissue structure and function. Advancing the “transcript-centric” view of scRNA-seq analyses is presently restricted by the limited resolution of proteomics and genome-wide techniques to analyze post-translational modifications. Here, by combining spatial cell sorting with transcriptomics and quantitative proteomics/phosphoproteomics, we established the spatially resolved proteome landscape of the liver endothelium, yielding deep mechanistic insight into zonated vascular signaling mechanisms. Phosphorylation of receptor tyrosine kinases was detected preferentially in the central vein area, resulting in an atypical enrichment of tyrosine phosphorylation. Prototypic biological validation identified Tie receptor signaling as a selective and specific regulator of vascular Wnt activity orchestrating angiocrine signaling, thereby controlling hepatocyte function during liver regeneration. Taken together, the study has yielded fundamental insight into the spatial organization of liver endothelial cell signaling. Spatial sorting may be employed as a universally adaptable strategy for multiomic analyses of scRNA-seq-defined cellular (sub)-populations. ScRNA-seq-guided spatial sort enables multiomic dissection of the liver vasculature Liver sinusoidal endothelial cells have a hybrid vascular-lymphatic phenotype Tyrosine phosphorylation of endothelial cell molecules is enriched on central vein Endothelial Tie1 shapes hepatic Wnt signal zonation and promotes liver regeneration
Collapse
Affiliation(s)
- Donato Inverso
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Jingjing Shi
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ki Hong Lee
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Moritz Jakab
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Shani Ben-Moshe
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shubhada R Kulkarni
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Martin Schneider
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guanxiong Wang
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Marziyeh Komeili
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Paula Argos Vélez
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Maria Riedel
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carleen Spegg
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Ruppert
- Center for Molecular Biology (ZMBH), Heidelberg University, Heidelberg, Germany
| | - Christine Schaeffer-Reiss
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Dominic Helm
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Indrabahadur Singh
- Emmy Noether Research Group Epigenetic Machineries and Cancer, Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center and Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | | | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Cancer Consortium, Heidelberg, Germany.
| |
Collapse
|
16
|
Browne S, Gill EL, Schultheiss P, Goswami I, Healy KE. Stem cell-based vascularization of microphysiological systems. Stem Cell Reports 2021; 16:2058-2075. [PMID: 33836144 PMCID: PMC8452487 DOI: 10.1016/j.stemcr.2021.03.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
Microphysiological systems (MPSs) (i.e., tissue or organ chips) exploit microfluidics and 3D cell culture to mimic tissue and organ-level physiology. The advent of human induced pluripotent stem cell (hiPSC) technology has accelerated the use of MPSs to study human disease in a range of organ systems. However, in the reduction of system complexity, the intricacies of vasculature are an often-overlooked aspect of MPS design. The growing library of pluripotent stem cell-derived endothelial cell and perivascular cell protocols have great potential to improve the physiological relevance of vasculature within MPS, specifically for in vitro disease modeling. Three strategic categories of vascular MPS are outlined: self-assembled, interface focused, and 3D biofabricated. This review discusses key features and development of the native vasculature, linking that to how hiPSC-derived vascular cells have been generated, the state of the art in vascular MPSs, and opportunities arising from interdisciplinary thinking.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Elisabeth L Gill
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Paula Schultheiss
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
17
|
Kennedy CC, Brown EE, Abutaleb NO, Truskey GA. Development and Application of Endothelial Cells Derived From Pluripotent Stem Cells in Microphysiological Systems Models. Front Cardiovasc Med 2021; 8:625016. [PMID: 33659279 PMCID: PMC7917070 DOI: 10.3389/fcvm.2021.625016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/11/2021] [Indexed: 12/02/2022] Open
Abstract
The vascular endothelium is present in all organs and blood vessels, facilitates the exchange of nutrients and waste throughout different organ systems in the body, and sets the tone for healthy vessel function. Mechanosensitive in nature, the endothelium responds to the magnitude and temporal waveform of shear stress in the vessels. Endothelial dysfunction can lead to atherosclerosis and other diseases. Modeling endothelial function and dysfunction in organ systems in vitro, such as the blood-brain barrier and tissue-engineered blood vessels, requires sourcing endothelial cells (ECs) for these biomedical engineering applications. It can be difficult to source primary, easily renewable ECs that possess the function or dysfunction in question. In contrast, human pluripotent stem cells (hPSCs) can be sourced from donors of interest and renewed almost indefinitely. In this review, we highlight how knowledge of vascular EC development in vivo is used to differentiate induced pluripotent stem cells (iPSC) into ECs. We then describe how iPSC-derived ECs are being used currently in in vitro models of organ function and disease and in vivo applications.
Collapse
Affiliation(s)
- Crystal C. Kennedy
- University Program in Genetics and Genomics, Duke University, Durham, NC, United States
| | - Erin E. Brown
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Nadia O. Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
18
|
Abstract
All organisms growing beyond the oxygen diffusion limit critically depend on a functional vasculature for survival. Yet blood vessels are far more than passive, uniform conduits for oxygen and nutrient supply. A remarkable organotypic heterogeneity is brought about by tissue-specific differentiated endothelial cells (lining the blood vessels' lumen) and allows blood vessels to deal with organ-specific demands for homeostasis. On the flip side, when blood vessels go awry, they promote life-threatening diseases characterized by endothelial cells inappropriately adopting an angiogenic state (eg, tumor vascularization) or becoming dysfunctional (eg, diabetic microvasculopathies), calling respectively for antiangiogenic therapies and proangiogenic/vascular regenerative strategies. In solid tumors, despite initial enthusiasm, growth factor-based (mostly anti-VEGF [vascular endothelial growth factor]) antiangiogenic therapies do not sufficiently live up to the expectations in terms of efficiency and patient survival, in part, due to intrinsic and acquired therapy resistance. Tumors cunningly deploy alternative growth factors than the ones targeted by the antiangiogenic therapies to reinstigate angiogenesis or revert to other ways of securing blood flow, independently of the targeted growth factors. In trying to alleviate tissue ischemia and to repair dysfunctional or damaged endothelium, local in-tissue administration of (genes encoding) proangiogenic factors or endothelial (stem) cells harnessing regenerative potential have been explored. Notwithstanding evaluation in clinical trials, these approaches are often hampered by dosing issues and limited half-life or local retention of the administered agents. Here, without intending to provide an all-encompassing historical overview, we focus on some recent advances in understanding endothelial cell behavior in health and disease and identify novel molecular players and concepts that could eventually be considered for therapeutic targeting.
Collapse
Affiliation(s)
- Guy Eelen
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Lucas Treps
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| | - Peter Carmeliet
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.).,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| |
Collapse
|
19
|
Blatchley MR, Gerecht S. Reconstructing the Vascular Developmental Milieu In Vitro. Trends Cell Biol 2020; 30:15-31. [DOI: 10.1016/j.tcb.2019.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
|
20
|
Rosa S, Praça C, Pitrez PR, Gouveia PJ, Aranguren XL, Ricotti L, Ferreira LS. Functional characterization of iPSC-derived arterial- and venous-like endothelial cells. Sci Rep 2019; 9:3826. [PMID: 30846769 PMCID: PMC6405900 DOI: 10.1038/s41598-019-40417-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/11/2019] [Indexed: 02/06/2023] Open
Abstract
The current work reports the functional characterization of human induced pluripotent stem cells (iPSCs)- arterial and venous-like endothelial cells (ECs), derived in chemically defined conditions, either in monoculture or seeded in a scaffold with mechanical properties similar to blood vessels. iPSC-derived arterial- and venous-like endothelial cells were obtained in two steps: differentiation of iPSCs into endothelial precursor cells (CD31pos/KDRpos/VE-Cadmed/EphB2neg/COUP-TFneg) followed by their differentiation into arterial and venous-like ECs using a high and low vascular endothelial growth factor (VEGF) concentration. Cells were characterized at gene, protein and functional levels. Functionally, both arterial and venous-like iPSC-derived ECs responded to vasoactive agonists such as thrombin and prostaglandin E2 (PGE2), similar to somatic ECs; however, arterial-like iPSC-derived ECs produced higher nitric oxide (NO) and elongation to shear stress than venous-like iPSC-derived ECs. Both cells adhered, proliferated and prevented platelet activation when seeded in poly(caprolactone) scaffolds. Interestingly, both iPSC-derived ECs cultured in monoculture or in a scaffold showed a different inflammatory profile than somatic ECs. Although both somatic and iPSC-derived ECs responded to tumor necrosis factor-α (TNF-α) by an increase in the expression of intercellular adhesion molecule 1 (ICAM-1), only somatic ECs showed an upregulation in the expression of E-selectin or vascular cell adhesion molecule 1 (VCAM-1).
Collapse
Affiliation(s)
- S Rosa
- CNC UC- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - C Praça
- CNC UC- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal
| | - P R Pitrez
- CNC UC- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal
| | - P José Gouveia
- CNC UC- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.,IIIUC- Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - X L Aranguren
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra, and Division of Oncology, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - L Ricotti
- The BioRobotics Institute, Scuola Superiore Sant' Anna, Viale Rinaldo Piaggio 34, 56025, Pontedera, PI, Italy
| | - L Silva Ferreira
- CNC UC- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal. .,Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal.
| |
Collapse
|