101
|
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: 33] [Impact Index Per Article: 8.3] [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
|
102
|
Betterman KL, Sutton DL, Secker GA, Kazenwadel J, Oszmiana A, Lim L, Miura N, Sorokin L, Hogan BM, Kahn ML, McNeill H, Harvey NL. Atypical cadherin FAT4 orchestrates lymphatic endothelial cell polarity in response to flow. J Clin Invest 2021; 130:3315-3328. [PMID: 32182215 DOI: 10.1172/jci99027] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/05/2020] [Indexed: 01/07/2023] Open
Abstract
The atypical cadherin FAT4 has established roles in the regulation of planar cell polarity and Hippo pathway signaling that are cell context dependent. The recent identification of FAT4 mutations in Hennekam syndrome, features of which include lymphedema, lymphangiectasia, and mental retardation, uncovered an important role for FAT4 in the lymphatic vasculature. Hennekam syndrome is also caused by mutations in collagen and calcium binding EGF domains 1 (CCBE1) and ADAM metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3), encoding a matrix protein and protease, respectively, that regulate activity of the key prolymphangiogenic VEGF-C/VEGFR3 signaling axis by facilitating the proteolytic cleavage and activation of VEGF-C. The fact that FAT4, CCBE1, and ADAMTS3 mutations underlie Hennekam syndrome suggested that all 3 genes might function in a common pathway. We identified FAT4 as a target gene of GATA-binding protein 2 (GATA2), a key transcriptional regulator of lymphatic vascular development and, in particular, lymphatic vessel valve development. Here, we demonstrate that FAT4 functions in a lymphatic endothelial cell-autonomous manner to control cell polarity in response to flow and is required for lymphatic vessel morphogenesis throughout development. Our data reveal a crucial role for FAT4 in lymphangiogenesis and shed light on the mechanistic basis by which FAT4 mutations underlie a human lymphedema syndrome.
Collapse
Affiliation(s)
- Kelly L Betterman
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Drew L Sutton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Genevieve A Secker
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Jan Kazenwadel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Anna Oszmiana
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Lillian Lim
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, University of Queensland, Saint Lucia, Queensland, Australia.,Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helen McNeill
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| |
Collapse
|
103
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
104
|
Krüger-Genge A, Hauser S, Neffe AT, Liu Y, Lendlein A, Pietzsch J, Jung F. Response of Endothelial Cells to Gelatin-Based Hydrogels. ACS Biomater Sci Eng 2021; 7:527-540. [PMID: 33496571 DOI: 10.1021/acsbiomaterials.0c01432] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The establishment of confluent endothelial cell (EC) monolayers on implanted materials has been identified as a concept to avoid thrombus formation but is a continuous challenge in cardiovascular device engineering. Here, material properties of gelatin-based hydrogels obtained by reacting gelatin with varying amounts of lysine diisocyanate ethyl ester were correlated with the functional state of hydrogel contacting venous EC (HUVEC) and HUVEC's ability to form a monolayer on these hydrogels. The density of adherent HUVEC on the softest hydrogel at 37 °C (G' = 1.02 kPa, E = 1.1 ± 0.3 kPa) was significantly lower (125 mm-1) than on the stiffer hydrogels (920 mm-1; G' = 2.515 and 5.02 kPa, E = 4.8 ± 0.8 and 10.3 ± 1.2 kPa). This was accompanied by increased matrix metalloprotease activity (9 pmol·min-2 compared to 0.6 pmol·min-2) and stress fiber formation, while cell-to-cell contacts were comparable. Likewise, release of eicosanoids (e.g., prostacyclin release of 1.7 vs 0.2 pg·mL-1·cell-1) and the pro-inflammatory cytokine MCP-1 (8 vs <1.5 pg·mL-1·cell-1) was higher on the softer than on the stiffer hydrogels. The expressions of pro-inflammatory markers COX-2, COX-1, and RAGE were slightly increased on all hydrogels on day 2 (up to 200% of the control), indicating a weak inflammation; however, the levels dropped to below the control from day 6. The study revealed that hydrogels with higher moduli approached the status of a functionally confluent HUVEC monolayer. The results indicate the promising potential especially of the discussed gelatin-based hydrogels with higher G' as biomaterials for implants foreseen for the venous system.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Axel T Neffe
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513 Teltow, Germany
| | - Yue Liu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513 Teltow, Germany.,Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328 Dresden, Germany.,School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany
| | - Friedrich Jung
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513 Teltow, Germany
| |
Collapse
|
105
|
Lee S, Kim S, Koo DJ, Yu J, Cho H, Lee H, Song JM, Kim SY, Min DH, Jeon NL. 3D Microfluidic Platform and Tumor Vascular Mapping for Evaluating Anti-Angiogenic RNAi-Based Nanomedicine. ACS NANO 2021; 15:338-350. [PMID: 33231435 DOI: 10.1021/acsnano.0c05110] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Three-dimensional (3D) visualization of tumor vasculature is a key factor in accurate evaluation of RNA interference (RNAi)-based antiangiogenic nanomedicine, a promising approach for cancer therapeutics. However, this remains challenging because there is not a physiologically relevant in vitro model or precise analytic methodology. To address this limitation, a strategy based on 3D microfluidic angiogenesis-on-a-chip and 3D tumor vascular mapping was developed for evaluating RNAi-based antiangiogenic nanomedicine. We developed a microfluidic model to recapitulate functional 3D angiogenic sprouting when co-cultured with various cancer cell types. This model enabled efficient and rapid assessment of antiangiogenic nanomedicine in treatment of hyper-angiogenic cancer. In addition, tissue-clearing-based whole vascular mapping of tumor xenograft allowed extraction of complex 3D morphological information in diverse quantitative parameters. Using this 3D imaging-based analysis, we observed tumor sub-regional differences in the antiangiogenic effect. Our systematic strategy can help in narrowing down the promising targets of antiangiogenic nanomedicine and then enables deep analysis of complex morphological changes in tumor vasculature, providing a powerful platform for the development of safe and effective nanomedicine for cancer therapeutics.
Collapse
Affiliation(s)
- Somin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seongchan Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Jun Koo
- Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea
| | - James Yu
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeongjun Cho
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyojin Lee
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5 Hwarangno 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Joon Myong Song
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung-Yon Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 08826, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
106
|
Placental blood flow sensing and regulation in fetal growth restriction. Placenta 2021; 113:23-28. [PMID: 33509641 PMCID: PMC8448138 DOI: 10.1016/j.placenta.2021.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
The mechanical force of blood flow is a fundamental determinant of vascular homeostasis. This frictional stimulation of cells, fluid shear stress (FSS), is increasingly recognised as being essential to placental development and function. Here, we focus on the role of FSS in regulating fetoplacental circulatory flow, both in normal pregnancy and that affected by fetal growth restriction (FGR). The fetus is reliant on placental perfusion to meet its circulatory and metabolic demands. Failure of normal vascular adaptation and the mechanisms enabling responsive interaction between fetoplacental and maternal circulations can result in FGR. FSS generates vasodilatation at least partly through the release of endothelial nitric oxide, a process thought to be vital for adequate blood flow. Where FGR is caused by placental dysfunction, placental vascular anatomy is altered, alongside endothelial dysfunction and hypoxia, each impacting upon the complex balance of FSS forces. Identifying specific mechanical sensors and the mechanisms governing how FSS force is converted into biochemical signals is a fast-paced area of research. Here, we raise awareness of Piezo1 proteins, recently discovered to be FSS-sensitive in fetoplacental endothelium, and with emerging roles in NO generation, vascular tone and angiogenesis. We discuss the emerging concept that activating mechanosensors such as Piezo1 ultimately results in the orchestrated processes of placental vascular adaptation. Piecing together the mechanisms governing endothelial responses to FSS in placental insufficiency is an important step towards developing new treatments for FGR.
Collapse
|
107
|
Ricard N, Bailly S, Guignabert C, Simons M. The quiescent endothelium: signalling pathways regulating organ-specific endothelial normalcy. Nat Rev Cardiol 2021; 18:565-580. [PMID: 33627876 PMCID: PMC7903932 DOI: 10.1038/s41569-021-00517-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Endothelial cells are at the interface between circulating blood and tissues. This position confers on them a crucial role in controlling oxygen and nutrient exchange and cellular trafficking between blood and the perfused organs. The endothelium adopts a structure that is specific to the needs and function of each tissue and organ and is subject to tissue-specific signalling input. In adults, endothelial cells are quiescent, meaning that they are not proliferating. Quiescence was considered to be a state in which endothelial cells are not stimulated but are instead slumbering and awaiting activating signals. However, new evidence shows that quiescent endothelium is fully awake, that it constantly receives and initiates functionally important signalling inputs and that this state is actively regulated. Signalling pathways involved in the maintenance of functionally quiescent endothelia are starting to be identified and are a combination of endocrine, autocrine, paracrine and mechanical inputs. The paracrine pathways confer a microenvironment on the endothelial cells that is specific to the perfused organs and tissues. In this Review, we present the current knowledge of organ-specific signalling pathways involved in the maintenance of endothelial quiescence and the pathologies associated with their disruption. Linking organ-specific pathways and human vascular pathologies will pave the way towards the development of innovative preventive strategies and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Nicolas Ricard
- grid.47100.320000000419368710Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Sabine Bailly
- grid.457348.9Université Grenoble Alpes, INSERM, CEA, BIG-Biologie du Cancer et de l’Infection, Grenoble, France
| | - Christophe Guignabert
- grid.414221.0INSERM UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Hôpital Marie Lannelongue, Le Plessis-Robinson, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Michael Simons
- grid.47100.320000000419368710Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA ,grid.47100.320000000419368710Department of Cell Biology, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
108
|
Luo J, Shi X, Lin Y, Yuan Y, Kural MH, Wang J, Ellis MW, Anderson CW, Zhang SM, Riaz M, Niklason LE, Qyang Y. Efficient Differentiation of Human Induced Pluripotent Stem Cells into Endothelial Cells under Xenogeneic-free Conditions for Vascular Tissue Engineering. Acta Biomater 2021; 119:184-196. [PMID: 33166710 PMCID: PMC8133308 DOI: 10.1016/j.actbio.2020.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/12/2020] [Accepted: 11/03/2020] [Indexed: 12/31/2022]
Abstract
Tissue engineered vascular grafts (TEVGs) represent a promising therapeutic option for emergency vascular intervention. Although the application of small-diameter TEVGs using patient-specific primary endothelial cells (ECs) to prevent thrombosis and occlusion prior to implantation could be hindered by the long time course required for in vitro endothelialization, human induced pluripotent stem cells (hiPSCs) provide a robust source to derive immunocompatible ECs (hiPSC-ECs) for immediate TEVG endothelialization. To achieve clinical application, hiPSC-ECs should be derived under culture conditions without the use of animal-derived reagents (xenogeneic-free conditions), to avoid unwanted host immune responses from xenogeneic reagents. However, a completely xenogeneic-free method of hiPSC-EC generation has not previously been established. Herein, we substituted animal-derived reagents used in a standard method of xenogeneic hiPSC-EC differentiation with functional counterparts of human origin. As a result, we generated xenogeneic-free hiPSC-ECs (XF-hiPSC-ECs) with similar marker expression and function to those of human primary ECs. Furthermore, XF-hiPSC-ECs functionally responded to shear stress with typical cell alignment and gene expression. Finally, we successfully endothelialized decellularized human vessels with XF-hiPSC-ECs in a dynamic bioreactor system. In conclusion, we developed xenogeneic-free conditions for generating functional hiPSC-ECs suitable for vascular tissue engineering, which will further move TEVG therapy toward clinical application.
Collapse
Affiliation(s)
- Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Xiangyu Shi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yuyao Lin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yifan Yuan
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA
| | - Mehmet H Kural
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA
| | - Juan Wang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA
| | - Matthew W Ellis
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06519, USA
| | - Christopher W Anderson
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, 06520 USA
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520 USA
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Laura E Niklason
- Yale Stem Cell Center, New Haven, CT 06520, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, 06520 USA.
| |
Collapse
|
109
|
Stassen OMJA, Ristori T, Sahlgren CM. Notch in mechanotransduction - from molecular mechanosensitivity to tissue mechanostasis. J Cell Sci 2020; 133:133/24/jcs250738. [PMID: 33443070 DOI: 10.1242/jcs.250738] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tissue development and homeostasis are controlled by mechanical cues. Perturbation of the mechanical equilibrium triggers restoration of mechanostasis through changes in cell behavior, while defects in these restorative mechanisms lead to mechanopathologies, for example, osteoporosis, myopathies, fibrosis or cardiovascular disease. Therefore, sensing mechanical cues and integrating them with the biomolecular cell fate machinery is essential for the maintenance of health. The Notch signaling pathway regulates cell and tissue fate in nearly all tissues. Notch activation is directly and indirectly mechanosensitive, and regulation of Notch signaling, and consequently cell fate, is integral to the cellular response to mechanical cues. Fully understanding the dynamic relationship between molecular signaling, tissue mechanics and tissue remodeling is challenging. To address this challenge, engineered microtissues and computational models play an increasingly large role. In this Review, we propose that Notch takes on the role of a 'mechanostat', maintaining the mechanical equilibrium of tissues. We discuss the reciprocal role of Notch in the regulation of tissue mechanics, with an emphasis on cardiovascular tissues, and the potential of computational and engineering approaches to unravel the complex dynamic relationship between mechanics and signaling in the maintenance of cell and tissue mechanostasis.
Collapse
Affiliation(s)
- Oscar M J A Stassen
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University, 20500 Turku, Finland.,Turku Bioscience Centre, Åbo Akademi University and University of Turku, 20520 Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Cecilia M Sahlgren
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University, 20500 Turku, Finland .,Turku Bioscience Centre, Åbo Akademi University and University of Turku, 20520 Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
110
|
Abstract
PURPOSE OF REVIEW Diabetes mellitus is a prevalent chronic disease affecting millions of people in the world. Bone fragility is a complication found in diabetic patients. Although osteoblasts and osteoclasts are directly affected by diabetes, herein we focus on how the diabetic state-based on hyperglycemia and accumulation of advanced glycation end products among other features-impairs osteocyte functions exerting deleterious effects on bone. RECENT FINDINGS In the last years, several studies described that diabetic conditions cause morphological modifications on lacunar-canalicular system, alterations on osteocyte mechanoreceptors and intracellular pathways and on osteocyte communication with other cells through the secretion of proteins such as sclerostin or RANKL. This article gives an overview of events occurring in diabetic osteocytes. In particular, mechanical responses seem to be seriously affected in these conditions, suggesting that mechanical sensibility could be a target for future research in the field.
Collapse
Affiliation(s)
- Arancha R Gortázar
- Bone Physiopathology laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925, Alcorcón, Madrid, Spain.
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU,CEU Universities, Campus Monteprincipe, 28925, Alcorcón, Madrid, Spain.
| | - Juan A Ardura
- Bone Physiopathology laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925, Alcorcón, Madrid, Spain
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU,CEU Universities, Campus Monteprincipe, 28925, Alcorcón, Madrid, Spain
| |
Collapse
|
111
|
Reina-Torres E, De Ieso ML, Pasquale LR, Madekurozwa M, van Batenburg-Sherwood J, Overby DR, Stamer WD. The vital role for nitric oxide in intraocular pressure homeostasis. Prog Retin Eye Res 2020; 83:100922. [PMID: 33253900 DOI: 10.1016/j.preteyeres.2020.100922] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Catalyzed by endothelial nitric oxide (NO) synthase (eNOS) activity, NO is a gaseous signaling molecule maintaining endothelial and cardiovascular homeostasis. Principally, NO regulates the contractility of vascular smooth muscle cells and permeability of endothelial cells in response to either biochemical or biomechanical cues. In the conventional outflow pathway of the eye, the smooth muscle-like trabecular meshwork (TM) cells and Schlemm's canal (SC) endothelium control aqueous humor outflow resistance, and therefore intraocular pressure (IOP). The mechanisms by which outflow resistance is regulated are complicated, but NO appears to be a key player as enhancement or inhibition of NO signaling dramatically affects outflow function; and polymorphisms in NOS3, the gene that encodes eNOS modifies the relation between various environmental exposures and glaucoma. Based upon a comprehensive review of past foundational studies, we present a model whereby NO controls a feedback signaling loop in the conventional outflow pathway that is sensitive to changes in IOP and its oscillations. Thus, upon IOP elevation, the outflow pathway tissues distend, and the SC lumen narrows resulting in increased SC endothelial shear stress and stretch. In response, SC cells upregulate the production of NO, relaxing neighboring TM cells and increasing permeability of SC's inner wall. These IOP-dependent changes in the outflow pathway tissues reduce the resistance to aqueous humor drainage and lower IOP, which, in turn, diminishes the biomechanical signaling on SC. Similar to cardiovascular pathogenesis, dysregulation of the eNOS/NO system leads to dysfunctional outflow regulation and ocular hypertension, eventually resulting in primary open-angle glaucoma.
Collapse
Affiliation(s)
| | | | - Louis R Pasquale
- Eye and Vision Research Institute of New York Eye and Ear Infirmary at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK.
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC, USA.
| |
Collapse
|
112
|
Shi X, He L, Zhang SM, Luo J. Human iPS Cell-derived Tissue Engineered Vascular Graft: Recent Advances and Future Directions. Stem Cell Rev Rep 2020; 17:862-877. [PMID: 33230612 DOI: 10.1007/s12015-020-10091-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 12/19/2022]
Abstract
Tissue engineered vascular grafts (TEVGs) generated from human primary cells represent a promising vascular interventional therapy. However, generation and application of these TEVGs may be significantly hindered by the limited accessibility, finite expandability, donor-donor functional variation and immune-incompatibility of primary seed cells from donors. Alternatively, human induced pluripotent stem cells (hiPSCs) offer an infinite source to obtain functional vascular cells in large quantity and comparable quality for TEVG construction. To date, TEVGs (hiPSC-TEVGs) with significant mechanical strength and implantability have been generated using hiPSC-derived seed cells. Despite being in its incipient stage, this emerging field of hiPSC-TEVG research has achieved significant progress and presented promising future potential. Meanwhile, a series of challenges pertaining hiPSC differentiation, vascular tissue engineering technologies and future production and application await to be addressed. Herein, we have composed this review to introduce progress in TEVG generation using hiPSCs, summarize the current major challenges, and encapsulate the future directions of research on hiPSC-based TEVGs. Graphical abstract.
Collapse
Affiliation(s)
- Xiangyu Shi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.,Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine , Yale School of Medicine, 300 George Street, Room 752, New Haven, CT, 06511, USA
| | - Lile He
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, 06520, New Haven, CT, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine , Yale School of Medicine, 300 George Street, Room 752, New Haven, CT, 06511, USA. .,Yale Stem Cell Center, 06520, New Haven, CT, USA.
| |
Collapse
|
113
|
Klems A, van Rijssel J, Ramms AS, Wild R, Hammer J, Merkel M, Derenbach L, Préau L, Hinkel R, Suarez-Martinez I, Schulte-Merker S, Vidal R, Sauer S, Kivelä R, Alitalo K, Kupatt C, van Buul JD, le Noble F. The GEF Trio controls endothelial cell size and arterial remodeling downstream of Vegf signaling in both zebrafish and cell models. Nat Commun 2020; 11:5319. [PMID: 33087700 PMCID: PMC7578835 DOI: 10.1038/s41467-020-19008-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Arterial networks enlarge in response to increase in tissue metabolism to facilitate flow and nutrient delivery. Typically, the transition of a growing artery with a small diameter into a large caliber artery with a sizeable diameter occurs upon the blood flow driven change in number and shape of endothelial cells lining the arterial lumen. Here, using zebrafish embryos and endothelial cell models, we describe an alternative, flow independent model, involving enlargement of arterial endothelial cells, which results in the formation of large diameter arteries. Endothelial enlargement requires the GEF1 domain of the guanine nucleotide exchange factor Trio and activation of Rho-GTPases Rac1 and RhoG in the cell periphery, inducing F-actin cytoskeleton remodeling, myosin based tension at junction regions and focal adhesions. Activation of Trio in developing arteries in vivo involves precise titration of the Vegf signaling strength in the arterial wall, which is controlled by the soluble Vegf receptor Flt1. Arterial flow regulates artery diameter but other mechanisms may also affect this. Here, the authors show that the guanine nucleotide exchange factor Trio and GTPases Rac1 and RhoG, triggers F-actin remodeling in arterial endothelial cells, independent of flow, to enhance lumen diameter in zebrafish and cell models.
Collapse
Affiliation(s)
- Alina Klems
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Jos van Rijssel
- Molecular Cell Biology lab, Department Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center at the University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Anne S Ramms
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany.,Institute for Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021, Karlsruhe, Germany
| | - Raphael Wild
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Julia Hammer
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Melanie Merkel
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Laura Derenbach
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Laetitia Préau
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany and DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Irina Suarez-Martinez
- Institute of Cardiovascular Organogenesis and Regeneration WWU Münster, Münster, Germany & Faculty of Medicine, WWU Münster, Münster, Germany & Cells in Motion Cluster of Excellence, Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration WWU Münster, Münster, Germany & Faculty of Medicine, WWU Münster, Münster, Germany & Cells in Motion Cluster of Excellence, Münster, Münster, Germany
| | - Ramon Vidal
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute of Medical Systems Biology & Berlin Institute of Health, Robert Rössle Strasse 10, 13092, Berlin, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute of Medical Systems Biology & Berlin Institute of Health, Robert Rössle Strasse 10, 13092, Berlin, Germany
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Helsinki, Finland
| | - Kari Alitalo
- Translational Cancer Medicine Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Helsinki, Finland
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, TUM Munich, Germany, and DZHK, (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jaap D van Buul
- Molecular Cell Biology lab, Department Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center at the University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, The Netherlands
| | - Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany. .,Institute for Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021, Karlsruhe, Germany. .,Institute of Experimental Cardiology, University of Heidelberg, Heidelberg Germany and DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
114
|
Morris G, Puri BK, Olive L, Carvalho A, Berk M, Walder K, Gustad LT, Maes M. Endothelial dysfunction in neuroprogressive disorders-causes and suggested treatments. BMC Med 2020; 18:305. [PMID: 33070778 PMCID: PMC7570030 DOI: 10.1186/s12916-020-01749-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Potential routes whereby systemic inflammation, oxidative stress and mitochondrial dysfunction may drive the development of endothelial dysfunction and atherosclerosis, even in an environment of low cholesterol, are examined. MAIN TEXT Key molecular players involved in the regulation of endothelial cell function are described, including PECAM-1, VE-cadherin, VEGFRs, SFK, Rho GEF TRIO, RAC-1, ITAM, SHP-2, MAPK/ERK, STAT-3, NF-κB, PI3K/AKT, eNOS, nitric oxide, miRNAs, KLF-4 and KLF-2. The key roles of platelet activation, xanthene oxidase and myeloperoxidase in the genesis of endothelial cell dysfunction and activation are detailed. The following roles of circulating reactive oxygen species (ROS), reactive nitrogen species and pro-inflammatory cytokines in the development of endothelial cell dysfunction are then described: paracrine signalling by circulating hydrogen peroxide, inhibition of eNOS and increased levels of mitochondrial ROS, including compromised mitochondrial dynamics, loss of calcium ion homeostasis and inactivation of SIRT-1-mediated signalling pathways. Next, loss of cellular redox homeostasis is considered, including further aspects of the roles of hydrogen peroxide signalling, the pathological consequences of elevated NF-κB, compromised S-nitrosylation and the development of hypernitrosylation and increased transcription of atherogenic miRNAs. These molecular aspects are then applied to neuroprogressive disorders by considering the following potential generators of endothelial dysfunction and activation in major depressive disorder, bipolar disorder and schizophrenia: NF-κB; platelet activation; atherogenic miRs; myeloperoxidase; xanthene oxidase and uric acid; and inflammation, oxidative stress, nitrosative stress and mitochondrial dysfunction. CONCLUSIONS Finally, on the basis of the above molecular mechanisms, details are given of potential treatment options for mitigating endothelial cell dysfunction and activation in neuroprogressive disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | | | - Lisa Olive
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- School of Psychology, Faculty of Health, Deakin University, Geelong, Australia
| | - Andre Carvalho
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia.
- Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Ken Walder
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Lise Tuset Gustad
- Department of Circulation and medical imaging, Norwegian University of Technology and Science (NTNU), Trondheim, Norway
- Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | - Michael Maes
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
115
|
Tsata V, Beis D. In Full Force. Mechanotransduction and Morphogenesis during Homeostasis and Tissue Regeneration. J Cardiovasc Dev Dis 2020; 7:jcdd7040040. [PMID: 33019569 PMCID: PMC7711708 DOI: 10.3390/jcdd7040040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022] Open
Abstract
The interactions of form and function have been the focus of numerous studies in the context of development and more recently regeneration. Our understanding on how cells, tissues and organs sense and interpret external cues, such as mechanical forces, is becoming deeper as novel techniques in imaging are applied and the relevant signaling pathways emerge. These cellular responses can be found from bacteria to all multicellular organisms such as plants and animals. In this review, we focus on hemodynamic flow and endothelial shear stress during cardiovascular development and regeneration, where the interactions of morphogenesis and proper function are more prominent. In addition, we address the recent literature on the role of extracellular matrix and fibrotic response during tissue repair and regeneration. Finally, we refer to examples where the integration of multi-disciplinary approaches to understand the biomechanics of cellular responses could be utilized in novel medical applications.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Correspondence: (V.T.); (D.B.); Tel.: +3021-0659-7439 (V.T. & D.B.)
| | - Dimitris Beis
- Correspondence: (V.T.); (D.B.); Tel.: +3021-0659-7439 (V.T. & D.B.)
| |
Collapse
|
116
|
Bosseboeuf E, Raimondi C. Signalling, Metabolic Pathways and Iron Homeostasis in Endothelial Cells in Health, Atherosclerosis and Alzheimer's Disease. Cells 2020; 9:cells9092055. [PMID: 32911833 PMCID: PMC7564205 DOI: 10.3390/cells9092055] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells drive the formation of new blood vessels in physiological and pathological contexts such as embryonic development, wound healing, cancer and ocular diseases. Once formed, all vessels of the vasculature system present an endothelial monolayer (the endothelium), lining the luminal wall of the vessels, that regulates gas and nutrient exchange between the circulating blood and tissues, contributing to maintaining tissue and vascular homeostasis. To perform their functions, endothelial cells integrate signalling pathways promoted by growth factors, cytokines, extracellular matrix components and signals from mechanosensory complexes sensing the blood flow. New evidence shows that endothelial cells rely on specific metabolic pathways for distinct cellular functions and that the integration of signalling and metabolic pathways regulates endothelial-dependent processes such as angiogenesis and vascular homeostasis. In this review, we provide an overview of endothelial functions and the recent advances in understanding the role of endothelial signalling and metabolism in physiological processes such as angiogenesis and vascular homeostasis and vascular diseases. Also, we focus on the signalling pathways promoted by the transmembrane protein Neuropilin-1 (NRP1) in endothelial cells, its recently discovered role in regulating mitochondrial function and iron homeostasis and the role of mitochondrial dysfunction and iron in atherosclerosis and neurodegenerative diseases.
Collapse
|
117
|
Douglas G, Mehta V, Al Haj Zen A, Akoumianakis I, Goel A, Rashbrook VS, Trelfa L, Donovan L, Drydale E, Chuaiphichai S, Antoniades C, Watkins H, Kyriakou T, Tzima E, Channon KM. A key role for the novel coronary artery disease gene JCAD in atherosclerosis via shear stress mechanotransduction. Cardiovasc Res 2020; 116:1863-1874. [PMID: 31584065 PMCID: PMC7449560 DOI: 10.1093/cvr/cvz263] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/13/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS Genome-wide association studies (GWAS) have consistently identified an association between coronary artery disease (CAD) and a locus on chromosome 10 containing a single gene, JCAD (formerly KIAA1462). However, little is known about the mechanism by which JCAD could influence the development of atherosclerosis. METHODS AND RESULTS Vascular function was quantified in subjects with CAD by flow-mediated dilatation (FMD) and vasorelaxation responses in isolated blood vessel segments. The JCAD risk allele identified by GWAS was associated with reduced FMD and reduced endothelial-dependent relaxations. To study the impact of loss of Jcad on atherosclerosis, Jcad-/- mice were crossed to an ApoE-/- background and fed a high-fat diet from 6 to16 weeks of age. Loss of Jcad did not affect blood pressure or heart rate. However, Jcad-/-ApoE-/- mice developed significantly less atherosclerosis in the aortic root and the inner curvature of the aortic arch. En face analysis revealed a striking reduction in pro-inflammatory adhesion molecules at sites of disturbed flow on the endothelial cell layer of Jcad-/- mice. Loss of Jcad lead to a reduced recovery perfusion in response to hind limb ischaemia, a model of altered in vivo flow. Knock down of JCAD using siRNA in primary human aortic endothelial cells significantly reduced the response to acute onset of flow, as evidenced by reduced phosphorylation of NF-КB, eNOS, and Akt. CONCLUSION The novel CAD gene JCAD promotes atherosclerotic plaque formation via a role in the endothelial cell shear stress mechanotransduction pathway.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/physiopathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/physiopathology
- Aortic Diseases/prevention & control
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/physiopathology
- Atherosclerosis/prevention & control
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cells, Cultured
- Coronary Artery Disease/genetics
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/physiopathology
- Coronary Circulation
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Disease Models, Animal
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Genome-Wide Association Study
- Hindlimb/blood supply
- Humans
- Ischemia/genetics
- Ischemia/metabolism
- Ischemia/physiopathology
- Male
- Mechanotransduction, Cellular
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- NF-kappa B/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Phosphorylation
- Plaque, Atherosclerotic
- Proto-Oncogene Proteins c-akt
- Stress, Mechanical
Collapse
Affiliation(s)
- Gillian Douglas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Vedanta Mehta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ayman Al Haj Zen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar Foundation, Doha, Qatar
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Victoria S Rashbrook
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lucy Trelfa
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lucy Donovan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Edward Drydale
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Surawee Chuaiphichai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Theodosios Kyriakou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ellie Tzima
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
118
|
Hiepen C, Mendez PL, Knaus P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020; 9:E1965. [PMID: 32858894 PMCID: PMC7564048 DOI: 10.3390/cells9091965] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGFβ) superfamily of cytokines. While some ligand members are potent inducers of angiogenesis, others promote vascular homeostasis. However, the precise understanding of the molecular mechanisms underlying these functions is still a growing research field. In bone, the tissue in which BMPs were first discovered, crosstalk of TGFβ/BMP signaling with mechanobiology is well understood. Likewise, the endothelium represents a tissue that is constantly exposed to multiple mechanical triggers, such as wall shear stress, elicited by blood flow or strain, and tension from the surrounding cells and to the extracellular matrix. To integrate mechanical stimuli, the cytoskeleton plays a pivotal role in the transduction of these forces in endothelial cells. Importantly, mechanical forces integrate on several levels of the TGFβ/BMP pathway, such as receptors and SMADs, but also global cell-architecture and nuclear chromatin re-organization. Here, we summarize the current literature on crosstalk mechanisms between biochemical cues elicited by TGFβ/BMP growth factors and mechanical cues, as shear stress or matrix stiffness that collectively orchestrate endothelial function. We focus on the different subcellular compartments in which the forces are sensed and integrated into the TGFβ/BMP growth factor signaling.
Collapse
Affiliation(s)
| | | | - Petra Knaus
- Knaus-Lab/Signal Transduction, Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (C.H.); (P.-L.M.)
| |
Collapse
|
119
|
Lymphatic Valves and Lymph Flow in Cancer-Related Lymphedema. Cancers (Basel) 2020; 12:cancers12082297. [PMID: 32824219 PMCID: PMC7464955 DOI: 10.3390/cancers12082297] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Lymphedema is a complex disease caused by the accumulation of fluid in the tissues resulting from a dysfunctional or damaged lymphatic vasculature. In developed countries, lymphedema most commonly occurs as a result of cancer treatment. Initially, impaired lymph flow causes edema, but over time this results in inflammation, fibrotic and fatty tissue deposition, limited mobility, and bacterial infections that can lead to sepsis. While chronically impaired lymph flow is generally believed to be the instigating factor, little is known about what pathophysiological changes occur in the lymphatic vessels to inhibit lymph flow. Lymphatic vessels not only regulate lymph flow through a variety of physiologic mechanisms, but also respond to lymph flow itself. One of the fascinating ways that lymphatic vessels respond to flow is by growing bicuspid valves that close to prevent the backward movement of lymph. However, lymphatic valves have not been investigated in cancer-related lymphedema patients, even though the mutations that cause congenital lymphedema regulate genes involved in valve development. Here, we review current knowledge of the regulation of lymphatic function and development by lymph flow, including newly identified genetic regulators of lymphatic valves, and provide evidence for lymphatic valve involvement in cancer-related lymphedema.
Collapse
|
120
|
Roux E, Bougaran P, Dufourcq P, Couffinhal T. Fluid Shear Stress Sensing by the Endothelial Layer. Front Physiol 2020; 11:861. [PMID: 32848833 PMCID: PMC7396610 DOI: 10.3389/fphys.2020.00861] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022] Open
Abstract
Blood flow produces mechanical frictional forces, parallel to the blood flow exerted on the endothelial wall of the vessel, the so-called wall shear stress (WSS). WSS sensing is associated with several vascular pathologies, but it is first a physiological phenomenon. Endothelial cell sensitivity to WSS is involved in several developmental and physiological vascular processes such as angiogenesis and vascular morphogenesis, vascular remodeling, and vascular tone. Local conditions of blood flow determine the characteristics of WSS, i.e., intensity, direction, pulsatility, sensed by the endothelial cells that, through their effect of the vascular network, impact WSS. All these processes generate a local-global retroactive loop that determines the ability of the vascular system to ensure the perfusion of the tissues. In order to account for the physiological role of WSS, the so-called shear stress set point theory has been proposed, according to which WSS sensing acts locally on vessel remodeling so that WSS is maintained close to a set point value, with local and distant effects of vascular blood flow. The aim of this article is (1) to review the existing literature on WSS sensing involvement on the behavior of endothelial cells and its short-term (vasoreactivity) and long-term (vascular morphogenesis and remodeling) effects on vascular functioning in physiological condition; (2) to present the various hypotheses about WSS sensors and analyze the conceptual background of these representations, in particular the concept of tensional prestress or biotensegrity; and (3) to analyze the relevance, explanatory value, and limitations of the WSS set point theory, that should be viewed as dynamical, and not algorithmic, processes, acting in a self-organized way. We conclude that this dynamic set point theory and the biotensegrity concept provide a relevant explanatory framework to analyze the physiological mechanisms of WSS sensing and their possible shift toward pathological situations.
Collapse
Affiliation(s)
- Etienne Roux
- Inserm, UMR 1034, Biology of Cardiovascular Diseases, University of Bordeaux, Bordeaux, France.,UMR 8560 IHPST - Institut d'Histoire et de Philosophie des Sciences et des Techniques, CNRS, Université Paris 1 Panthéon-Sorbonne, Paris, France
| | - Pauline Bougaran
- Inserm, UMR 1034, Biology of Cardiovascular Diseases, University of Bordeaux, Bordeaux, France
| | - Pascale Dufourcq
- Inserm, UMR 1034, Biology of Cardiovascular Diseases, University of Bordeaux, Bordeaux, France
| | - Thierry Couffinhal
- Inserm, UMR 1034, Biology of Cardiovascular Diseases, University of Bordeaux, Bordeaux, France
| |
Collapse
|
121
|
Paul MD, Grubb HN, Hristova K. Quantifying the strength of heterointeractions among receptor tyrosine kinases from different subfamilies: Implications for cell signaling. J Biol Chem 2020; 295:9917-9933. [PMID: 32467228 PMCID: PMC7380177 DOI: 10.1074/jbc.ra120.013639] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/20/2020] [Indexed: 01/09/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) are single-pass membrane proteins that control vital cell processes such as cell growth, survival, and differentiation. There is a growing body of evidence that RTKs from different subfamilies can interact and that these diverse interactions can have important biological consequences. However, these heterointeractions are often ignored, and their strengths are unknown. In this work, we studied the heterointeractions of nine RTK pairs, epidermal growth factor receptor (EGFR)-EPH receptor A2 (EPHA2), EGFR-vascular endothelial growth factor receptor 2 (VEGFR2), EPHA2-VEGFR2, EPHA2-fibroblast growth factor receptor 1 (FGFR1), EPHA2-FGFR2, EPHA2-FGFR3, VEGFR2-FGFR1, VEGFR2-FGFR2, and VEGFR2-FGFR3, using a FRET-based method. Surprisingly, we found that RTK heterodimerization and homodimerization strengths can be similar, underscoring the significance of RTK heterointeractions in signaling. We discuss how these heterointeractions can contribute to the complexity of RTK signal transduction, and we highlight the utility of quantitative FRET for probing multiple interactions in the plasma membrane.
Collapse
Affiliation(s)
- Michael D Paul
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hana N Grubb
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kalina Hristova
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
122
|
Gordon E, Schimmel L, Frye M. The Importance of Mechanical Forces for in vitro Endothelial Cell Biology. Front Physiol 2020; 11:684. [PMID: 32625119 PMCID: PMC7314997 DOI: 10.3389/fphys.2020.00684] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Blood and lymphatic vessels are lined by endothelial cells which constantly interact with their luminal and abluminal extracellular environments. These interactions confer physical forces on the endothelium, such as shear stress, stretch and stiffness, to mediate biological responses. These physical forces are often altered during disease, driving abnormal endothelial cell behavior and pathology. Therefore, it is critical that we understand the mechanisms by which endothelial cells respond to physical forces. Traditionally, endothelial cells in culture are grown in the absence of flow on stiff substrates such as plastic or glass. These cells are not subjected to the physical forces that endothelial cells endure in vivo, thus the results of these experiments often do not mimic those observed in the body. The field of vascular biology now realize that an intricate analysis of endothelial signaling mechanisms requires complex in vitro systems to mimic in vivo conditions. Here, we will review what is known about the mechanical forces that guide endothelial cell behavior and then discuss the advancements in endothelial cell culture models designed to better mimic the in vivo vascular microenvironment. A wider application of these technologies will provide more biologically relevant information from cultured cells which will be reproducible to conditions found in the body.
Collapse
Affiliation(s)
- Emma Gordon
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Lilian Schimmel
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
123
|
Pawlak JB, Bálint L, Lim L, Ma W, Davis RB, Benyó Z, Soares MJ, Oliver G, Kahn ML, Jakus Z, Caron KM. Lymphatic mimicry in maternal endothelial cells promotes placental spiral artery remodeling. J Clin Invest 2020; 129:4912-4921. [PMID: 31415243 DOI: 10.1172/jci120446] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/08/2019] [Indexed: 12/27/2022] Open
Abstract
Molecular heterogeneity of endothelial cells underlies their highly specialized functions during changing physiological conditions within diverse vascular beds. For example, placental spiral arteries (SAs) undergo remarkable remodeling to meet the ever-growing demands of the fetus - a process which is deficient in preeclampsia. The extent to which maternal endothelial cells coordinate with immune cells and pregnancy hormones to promote SA remodeling remains largely unknown. Here we found that remodeled SAs expressed the lymphatic markers PROX1, LYVE1, and VEGFR3, mimicking lymphatic identity. Uterine natural killer (uNK) cells, which are required for SA remodeling and secrete VEGFC, were both sufficient and necessary for VEGFR3 activation in vitro and in mice lacking uNK cells, respectively. Using Flt4Chy/+ mice with kinase inactive VEGFR3 and Vegfcfl/fl Vav1-Cre mice, we demonstrated that SA remodeling required VEGFR3 signaling, and that disrupted maternal VEGFR3 signaling contributed to late-gestation fetal growth restriction. Collectively, we identified a novel instance of lymphatic mimicry by which maternal endothelial cells promote SA remodeling, furthering our understanding of the vascular heterogeneity employed for the mitigation of pregnancy complications such as fetal growth restriction and preeclampsia.
Collapse
Affiliation(s)
- John B Pawlak
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - László Bálint
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary.,MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Lillian Lim
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wanshu Ma
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Reema B Davis
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Zoltán Benyó
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Michael J Soares
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Center for Perinatal Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri, USA
| | - Guillermo Oliver
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary.,MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
124
|
Norden PR, Sabine A, Wang Y, Demir CS, Liu T, Petrova TV, Kume T. Shear stimulation of FOXC1 and FOXC2 differentially regulates cytoskeletal activity during lymphatic valve maturation. eLife 2020; 9:53814. [PMID: 32510325 PMCID: PMC7302880 DOI: 10.7554/elife.53814] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 06/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in the transcription factor FOXC2 are predominately associated with lymphedema. Herein, we demonstrate a key role for related factor FOXC1, in addition to FOXC2, in regulating cytoskeletal activity in lymphatic valves. FOXC1 is induced by laminar, but not oscillatory, shear and inducible, endothelial-specific deletion impaired postnatal lymphatic valve maturation in mice. However, deletion of Foxc2 induced valve degeneration, which is exacerbated in Foxc1; Foxc2 mutants. FOXC1 knockdown (KD) in human lymphatic endothelial cells increased focal adhesions and actin stress fibers whereas FOXC2-KD increased focal adherens and disrupted cell junctions, mediated by increased ROCK activation. ROCK inhibition rescued cytoskeletal or junctional integrity changes induced by inactivation of FOXC1 and FOXC2 invitro and vivo respectively, but only ameliorated valve degeneration in Foxc2 mutants. These results identify both FOXC1 and FOXC2 as mediators of mechanotransduction in the postnatal lymphatic vasculature and posit cytoskeletal signaling as a therapeutic target in lymphatic pathologies.
Collapse
Affiliation(s)
- Pieter R Norden
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Amélie Sabine
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, United States
| | - Cansaran Saygili Demir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Ting Liu
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Tatiana V Petrova
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| |
Collapse
|
125
|
Zhang F, Zarkada G, Yi S, Eichmann A. Lymphatic Endothelial Cell Junctions: Molecular Regulation in Physiology and Diseases. Front Physiol 2020; 11:509. [PMID: 32547411 PMCID: PMC7274196 DOI: 10.3389/fphys.2020.00509] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Lymphatic endothelial cells (LECs) lining lymphatic vessels develop specialized cell-cell junctions that are crucial for the maintenance of vessel integrity and proper lymphatic vascular functions. Successful lymphatic drainage requires a division of labor between lymphatic capillaries that take up lymph via open "button-like" junctions, and collectors that transport lymph to veins, which have tight "zipper-like" junctions that prevent lymph leakage. In recent years, progress has been made in the understanding of these specialized junctions, as a result of the application of state-of-the-art imaging tools and novel transgenic animal models. In this review, we discuss lymphatic development and mechanisms governing junction remodeling between button and zipper-like states in LECs. Understanding lymphatic junction remodeling is important in order to unravel lymphatic drainage regulation in obesity and inflammatory diseases and may pave the way towards future novel therapeutic interventions.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Georgia Zarkada
- Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Sanjun Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale School of Medicine, Yale University, New Haven, CT, United States.,INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
126
|
Schimmel L, Fukuhara D, Richards M, Jin Y, Essebier P, Frampton E, Hedlund M, Dejana E, Claesson-Welsh L, Gordon E. c-Src controls stability of sprouting blood vessels in the developing retina independently of cell-cell adhesion through focal adhesion assembly. Development 2020; 147:dev185405. [PMID: 32108024 PMCID: PMC7157583 DOI: 10.1242/dev.185405] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/19/2020] [Indexed: 12/22/2022]
Abstract
Endothelial cell adhesion is implicated in blood vessel sprout formation, yet how adhesion controls angiogenesis, and whether it occurs via rapid remodeling of adherens junctions or focal adhesion assembly, or both, remains poorly understood. Furthermore, how endothelial cell adhesion is controlled in particular tissues and under different conditions remains unexplored. Here, we have identified an unexpected role for spatiotemporal c-Src activity in sprouting angiogenesis in the retina, which is in contrast to the dominant focus on the role of c-Src in the maintenance of vascular integrity. Thus, mice specifically deficient in endothelial c-Src displayed significantly reduced blood vessel sprouting and loss in actin-rich filopodial protrusions at the vascular front of the developing retina. In contrast to what has been observed during vascular leakage, endothelial cell-cell adhesion was unaffected by loss of c-Src. Instead, decreased angiogenic sprouting was due to loss of focal adhesion assembly and cell-matrix adhesion, resulting in loss of sprout stability. These results demonstrate that c-Src signaling at specified endothelial cell membrane compartments (adherens junctions or focal adhesions) control vascular processes in a tissue- and context-dependent manner.
Collapse
Affiliation(s)
- Lilian Schimmel
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Daisuke Fukuhara
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| | - Mark Richards
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| | - Yi Jin
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| | - Patricia Essebier
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Emmanuelle Frampton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Marie Hedlund
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| | - Elisabetta Dejana
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| | - Lena Claesson-Welsh
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| | - Emma Gordon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Uppsala University, Beijer and Science for Life Laboratories, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala 75185, Sweden
| |
Collapse
|
127
|
Sedlak JM, Clyne AM. A Modified Parallel Plate Flow Chamber to Study Local Endothelial Response to Recirculating Disturbed Flow. J Biomech Eng 2020; 142:041003. [PMID: 31536122 PMCID: PMC7104763 DOI: 10.1115/1.4044899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 08/28/2019] [Indexed: 11/08/2022]
Abstract
Atherosclerosis develops at arterial sites where endothelial cells (ECs) are exposed to low time-averaged shear stress, in particular in regions of recirculating disturbed flow. To understand how hemodynamics contributes to EC dysfunction in atheroma development, an in vitro parallel plate flow chamber gasket was modified with protruding baffles to produce large recirculating flow regions. Computational fluid dynamics (CFD) predicted that more than 60% of the flow surface area was below the 12 dynes/cm2 atheroprotective threshold. Bovine aortic endothelial cells (BAECs) were then seeded in the parallel plate flow chamber with either the standard laminar or the new disturbed flow gasket (DFG) and exposed to flow for 36 h. Cell morphology, nitric oxide (NO), proliferation, permeability, and monocyte adhesion were assessed by phase contrast and confocal microscopy. BAEC exposed to 20 dynes/cm2 shear stress in the laminar flow device aligned and elongated in the flow direction while increasing nitric oxide, decreasing permeability, and maintaining low proliferation and monocyte adhesion. BAEC in the recirculating flow and low shear stress disturbed flow device regions did not elongate or align, produced less nitric oxide, and showed higher proliferation, permeability, and monocyte adhesion than cells in the laminar flow device. However, cells in disturbed flow device regions exposed to atheroprotective shear stress did not consistently align or decrease permeability, and these cells demonstrated low nitric oxide levels. The new parallel plate DFG provides a means to study recirculating flow, highlighting the complex relationship between hemodynamics and endothelial function.
Collapse
Affiliation(s)
- Jason Matthew Sedlak
- School of Biomedical Engineering, Science, and Health Systems,
Drexel University, 3141 Chestnut Street,
Philadelphia, PA 19104
e-mail:
| | - Alisa Morss Clyne
- Fellow ASME Department of Mechanical Engineering, Drexel
University, 3141 Chestnut Street, Philadelphia,
PA 19104 e-mail:
| |
Collapse
|
128
|
It takes more than two to tango: mechanosignaling of the endothelial surface. Pflugers Arch 2020; 472:419-433. [PMID: 32239285 PMCID: PMC7165135 DOI: 10.1007/s00424-020-02369-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
The endothelial surface is a highly flexible signaling hub which is able to sense the hemodynamic forces of the streaming blood. The subsequent mechanosignaling is basically mediated by specific structures, like the endothelial glycocalyx building the top surface layer of endothelial cells as well as mechanosensitive ion channels within the endothelial plasma membrane. The mechanical properties of the endothelial cell surface are characterized by the dynamics of cytoskeletal proteins and play a key role in the process of signal transmission from the outside (lumen of the blood vessel) to the interior of the cell. Thus, the cell mechanics directly interact with the function of mechanosensitive structures and ion channels. To precisely maintain the vascular tone, a coordinated functional interdependency between endothelial cells and vascular smooth muscle cells is necessary. This is given by the fact that mechanosensitive ion channels are expressed in both cell types and that signals are transmitted via autocrine/paracrine mechanisms from layer to layer. Thus, the outer layer of the endothelial cells can be seen as important functional mechanosensitive and reactive cellular compartment. This review aims to describe the known mechanosensitive structures of the vessel building a bridge between the important role of physiological mechanosignaling and the proper vascular function. Since mutations and dysfunction of mechanosensitive proteins are linked to vascular pathologies such as hypertension, they play a potent role in the field of channelopathies and mechanomedicine.
Collapse
|
129
|
Mtshali Z, Moodley J, Naicker T. An Insight into the Angiogenic and Lymphatic Interplay in Pre-eclampsia Comorbid with HIV Infection. Curr Hypertens Rep 2020; 22:35. [PMID: 32200445 DOI: 10.1007/s11906-020-01040-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW To provide insight on the imbalance of angiogenic and lymphangiogenic factors in pre-eclampsia, as well as highlight polymorphism in genes related to angiogenesis and lymphangiogenesis. RECENT FINDINGS The pregnancy-specific disorder pre-eclampsia is diagnosed by the presence of hypertension with/without proteinuria, after 20 weeks of gestation. The pathogenesis of pre-eclampsia remains ambiguous, but research over the years has identified an imbalance in maternal and foetal factors. Familial predisposition and gene variation are also linked to pre-eclampsia development. The sFlt-1/PIGF ratio has attracted great attention over the years; more recently several researchers have reported that a sFlt-1/PIGF ratio of ≤ 38 can be used to predict short-term absence of pre-eclampsia. This ratio has the potential to prevent adverse pregnancy outcomes and reduce healthcare costs significantly. Genome-wide studies have additionally identified variation in the foetal gene near Flt-1. The development of preeclampsia is not limited to the maternal interface, but foetal involvement as well as genetic interplay is associated with the disorder.
Collapse
Affiliation(s)
- Zamahlabangane Mtshali
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.
| | - Jagidesa Moodley
- Department of Obstetrics and Gynaecology and Women's Health and HIV Research Group, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
130
|
Putative Receptors for Gravity Sensing in Mammalian Cells: The Effects of Microgravity. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10062028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gravity is a constitutive force that influences life on Earth. It is sensed and translated into biochemical stimuli through the so called “mechanosensors”, proteins able to change their molecular conformation in order to amplify external cues causing several intracellular responses. Mechanosensors are widely represented in the human body with important structures such as otholiths in hair cells of vestibular system and statoliths in plants. Moreover, they are also present in the bone, where mechanical cues can cause bone resorption or formation and in muscle in which mechanical stimuli can increase the sensibility for mechanical stretch. In this review, we discuss the role of mechanosensors in two different conditions: normogravity and microgravity, emphasizing their emerging role in microgravity. Microgravity is a singular condition in which many molecular changes occur, strictly connected with the modified gravity force and free fall of bodies. Here, we first summarize the most important mechanosensors involved in normogravity and microgravity. Subsequently, we propose muscle LIM protein (MLP) and sirtuins as new actors in mechanosensing and signaling transduction under microgravity.
Collapse
|
131
|
Tian YI, Zhang X, Torrejon K, Danias J, Gindina S, Nayyar A, Du Y, Xie Y. A bioengineering approach to Schlemm's canal-like stem cell differentiation for in vitro glaucoma drug screening. Acta Biomater 2020; 105:203-213. [PMID: 31982588 DOI: 10.1016/j.actbio.2020.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/30/2022]
Abstract
Human Schlemm's canal (HSC) cells are critical for understanding outflow physiology and glaucoma etiology. However, primary donor cells frequently used in research are difficult to isolate. HSC cells exhibit both vascular and lymphatic markers. Human adipose-derived stem cells (ADSCs) represent a potential source of HSC due to their capacity to differentiate into both vascular and lymphatic endothelial cells, via VEGF-A and VEGF-C. Shear stress plays a critical role in maintaining HSC integrity, function, and PROX1 expression. Additionally, the human trabecular meshwork (HTM) microenvironment could provide cues for HSC-like differentiation. We hypothesize that subjecting ADSCs to VEGF-A or VEGF-C, shear stress, and co-culture with HTM cells could provide biological, mechanical, and cellular cues necessary for HSC-like differentiation. To test this hypothesis, effects of VEGF-A, VEGF-C, and shear stress on ADSC differentiation were examined and compared to primary HSC cells in terms of cell morphology, and HSC marker expression using qPCR, immunoblotting, and immunocytochemistry analysis. Furthermore, the effect of co-culture with HTM cells on porous scaffolds on ADSC differentiation was studied. Treatment with VEGF-C under shear stress is effective in differentiating ADSCs into PROX1-expressing HSC-like cells. Co-culture with HTM cells on porous scaffolds leads to HTM/ADSC-derived HSC-like constructs that regulate through-flow and respond as expected to dexamethasone. STATEMENT OF SIGNIFICANCE: We successfully generated human Schlemm's canal (HSC) like cells from adipocyte-derived stem cells induced by biochemical and biomechanical cues as well as bioengineered human trabecular meshwork (HTM) on micropatterned, porous SU8 scaffolds. These stem cell-derived HSC-like cells co-cultured with HTM cells on SU8 scaffolds can regulate through-flow, and in particular, are responsive to steroid treatment as expected. These findings show that ADSC-derived HSC-like cells have the potential to recreate the ocular outflow pathway for in vitro glaucoma drug screening. To the best of our knowledge, it is the very first time to demonstrate derivation of Schlemm's canal-like cells from stem cells. It provides an important alternative source to primary Schlemm's canal cells that are very difficult to be isolated and cultured from human donors.
Collapse
Affiliation(s)
- Yangzi Isabel Tian
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - Xulang Zhang
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - Karen Torrejon
- Glauconix Biosciences, Inc., 251 Fuller Road, Albany, NY 12203, USA
| | - John Danias
- SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Sofya Gindina
- SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Ashima Nayyar
- SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Yiqin Du
- University of Pittsburg School of Medicine, 203 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Yubing Xie
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA.
| |
Collapse
|
132
|
Perpendicular alignment of lymphatic endothelial cells in response to spatial gradients in wall shear stress. Commun Biol 2020; 3:57. [PMID: 32029852 PMCID: PMC7005002 DOI: 10.1038/s42003-019-0732-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/27/2019] [Indexed: 02/05/2023] Open
Abstract
One-way valves in the lymphatic system form from lymphatic endothelial cells (LECs) during embryonic development and are required for efficient tissue drainage. Although fluid flow is thought to guide both valve formation and maintenance, how this occurs at a mechanistic level remains incompletely understood. We built microfluidic devices that reproduce critical aspects of the fluid flow patterns found at sites of valvulogenesis. Using these devices, we observed that LECs replicated aspects of the early steps in valvulogenesis: cells oriented perpendicular to flow in the region of maximum wall shear stress (WSS) and exhibited enhanced nuclear localization of FOXC2, a transcription factor required for valvulogenesis. Further experiments revealed that the cell surface protein E-selectin was required for both of these responses. Our observations suggest that spatial gradients in WSS help to demarcate the locations of valve formation, and implicate E-selectin as a component of a mechanosensory process for detecting WSS gradients. Using microfluidic systems, Michalaki et al show that lymphatic endothelial cells exposed to wall shear stress orient in the direction perpendicular to flow and show increased nuclear FOXC2 levels in a manner dependent on E-selectin, a transmembrane adhesion protein. These data provide insights into how lymphatic vessels respond to local flow-mediated mechanical cues.
Collapse
|
133
|
Liu Y, Lei P, Row S, Andreadis ST. Cadherin-11 binds to PDGFRβ and enhances cell proliferation and tissue regeneration via the PDGFR-AKT signaling axis. FASEB J 2020; 34:3792-3804. [PMID: 31930567 DOI: 10.1096/fj.201902613r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 11/11/2022]
Abstract
Intercellular adhesion through homotypic interaction between cadherins regulates multiple cellular processes including cytoskeletal organization, proliferation, and survival. In this paper, we provide evidence that cadherin-11 (CDH11) binds to and promotes cell proliferation both in vitro and in vivo in synergy with the platelet-derived growth factor receptor beta (PDGFRβ). Engagement of CDH11 increased the sensitivity of cells to PDGF-BB by 10- to 100-fold, resulting in rapid and sustained phosphorylation of AKT, ultimately promoting and cell proliferation and tissue regeneration. Indeed, wound healing experiments showed that healing was severely compromised in Cdh11-/- mice, as evidenced by significantly decreased proliferation, AKT phosphorylation, and extracellular matrix (ECM) synthesis of dermal cells. Our results shed light into understanding how intercellular adhesion can promote cell proliferation and may have implications for tissue regeneration and cancer progression.
Collapse
Affiliation(s)
- Yayu Liu
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, NY
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY
| | - Sindhu Row
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY
| | - Stelios T Andreadis
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, NY.,Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY.,Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY
| |
Collapse
|
134
|
Smith Q, Macklin B, Chan XY, Jones H, Trempel M, Yoder MC, Gerecht S. Differential HDAC6 Activity Modulates Ciliogenesis and Subsequent Mechanosensing of Endothelial Cells Derived from Pluripotent Stem Cells. Cell Rep 2020; 24:895-908.e6. [PMID: 30044986 DOI: 10.1016/j.celrep.2018.06.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/30/2018] [Accepted: 06/20/2018] [Indexed: 01/10/2023] Open
Abstract
The role of primary cilia in mechanosensation is essential in endothelial cell (EC) shear responsiveness. Here, we find that venous, capillary, and progenitor ECs respond to shear stress in vitro in a cilia-dependent manner. We then demonstrate that primary cilia assembly in human induced pluripotent stem cell (hiPSC)-derived ECs varies between different cell lines with marginal influence of differentiation protocol. hiPSC-derived ECs lacking cilia do not align to shear stress, lack stress fiber assembly, have uncoordinated migration during wound closure in vitro, and have aberrant calcium influx upon shear exposure. Transcriptional analysis reveals variation in regulatory genes involved in ciliogenesis among different hiPSC-derived ECs. Moreover, inhibition of histone deacetylase 6 (HDAC6) activity in hiPSC-ECs lacking cilia rescues cilia formation and restores mechanical sensing. Taken together, these results show the importance of primary cilia in hiPSC-EC mechano-responsiveness and its modulation through HDAC6 activity varies among hiPSC-ECs.
Collapse
Affiliation(s)
- Quinton Smith
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bria Macklin
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xin Yi Chan
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hannah Jones
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michelle Trempel
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Mervin C Yoder
- Department of Pediatrics, Biochemistry, and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
135
|
Cong X, Zhang SM, Batty L, Luo J. Application of Human Induced Pluripotent Stem Cells in Generating Tissue-Engineered Blood Vessels as Vascular Grafts. Stem Cells Dev 2019; 28:1581-1594. [PMID: 31663439 DOI: 10.1089/scd.2019.0234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In pace with the advancement of tissue engineering during recent decades, tissue-engineered blood vessels (TEBVs) have been generated using primary seed cells, and their impressive success in clinical trials have demonstrated the great potential of these TEBVs as implantable vascular grafts in human regenerative medicine. However, the production, therapeutic efficacy, and readiness in emergencies of current TEBVs could be hindered by the accessibility, expandability, and donor-donor variation of patient-specific primary seed cells. Alternatively, using human induced pluripotent stem cells (hiPSCs) to derive seed vascular cells for vascular tissue engineering could fundamentally address this current dilemma in TEBV production. As an emerging research field with a promising future, the generation of hiPSC-based TEBVs has been reported recently with significant progress. Simultaneously, to further promote hiPSC-based TEBVs into vascular grafts for clinical use, several challenges related to the safety, readiness, and structural integrity of vascular tissue need to be addressed. Herein, this review will focus on the evolution and role of hiPSCs in vascular tissue engineering technology and summarize the current progress, challenges, and future directions of research on hiPSC-based TEBVs.
Collapse
Affiliation(s)
- Xiaoqiang Cong
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cardiology, Bethune First Hospital of Jilin University, ChangChun, China
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Luke Batty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Yale Stem Cell Center, School of Medicine, Yale University, New Haven, Connecticut
| |
Collapse
|
136
|
Paddillaya N, Mishra A, Kondaiah P, Pullarkat P, Menon GI, Gundiah N. Biophysics of Cell-Substrate Interactions Under Shear. Front Cell Dev Biol 2019; 7:251. [PMID: 31781558 PMCID: PMC6857480 DOI: 10.3389/fcell.2019.00251] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/10/2019] [Indexed: 12/31/2022] Open
Abstract
Cells adhere to substrates through mechanosensitive focal adhesion complexes. Measurements that probe how cells detach from substrates when they experience an applied force connect molecular-scale aspects of cell adhesion with the biophysical properties of adherent cells. Such forces can be applied through shear devices that flow fluid in a controlled manner across cells. The signaling pathways associated with focal adhesions, in particular those that involve integrins and receptor tyrosine kinases, are complex, receiving mechano-chemical feedback from the sensing of substrate stiffness as well as of external forces. This article reviews the signaling processes involved in mechanosensing and mechanotransduction during cell-substrate interactions, describing the role such signaling plays in cancer metastasis. We examine some recent progress in quantifying the strength of these interactions, describing a novel fluid shear device that allows for the visualization of the cell and its sub-cellular structures under a shear flow. We also summarize related results from a biophysical model for cellular de-adhesion induced by applied forces. Quantifying cell-substrate adhesions under shear should aid in the development of mechano-diagnostic techniques for diseases in which cell-adhesion is mis-regulated, such as cancers.
Collapse
Affiliation(s)
- Neha Paddillaya
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Ashish Mishra
- Soft Condensed Matter Group, Raman Research Institute, Bangalore, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Pramod Pullarkat
- Soft Condensed Matter Group, Raman Research Institute, Bangalore, India
| | - Gautam I Menon
- The Institute of Mathematical Sciences, Chennai, India.,Homi Bhabha National Institute, Mumbai, India.,Department of Physics, Ashoka University, Sonepat, India
| | - Namrata Gundiah
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India.,Department of Mechanical Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
137
|
Ansari A, Schultheis K, Patel R, Al‐Qadi KI, Chen S, Jensen CR, Schad SR, Weddell JC, Vanka SP, Imoukhuede PI. Cell isolation via spiral microfluidics and the secondary anchor targeted cell release system. AIChE J 2019. [DOI: 10.1002/aic.16844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Ali Ansari
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Kinsey Schultheis
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Reema Patel
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Kareem I. Al‐Qadi
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Si Chen
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Cassandra R. Jensen
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Samantha R. Schad
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Jared C. Weddell
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - Surya P. Vanka
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| | - P. I. Imoukhuede
- Bioengineering University of Illinois at Urbana‐Champaign Champaign Illinois
| |
Collapse
|
138
|
Duchemin AL, Vignes H, Vermot J, Chow R. Mechanotransduction in cardiovascular morphogenesis and tissue engineering. Curr Opin Genet Dev 2019; 57:106-116. [PMID: 31586750 DOI: 10.1016/j.gde.2019.08.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/06/2019] [Accepted: 08/10/2019] [Indexed: 12/13/2022]
Abstract
Cardiovascular morphogenesis involves cell behavior and cell identity changes that are activated by mechanical forces associated with heart function. Recently, advances in in vivo imaging, methods to alter blood flow, and computational modelling have greatly advanced our understanding of how forces produced by heart contraction and blood flow impact different morphogenetic processes. Meanwhile, traditional genetic approaches have helped to elucidate how endothelial cells respond to forces at the cellular and molecular level. Here we discuss the principles of endothelial mechanosensitity and their interplay with cellular processes during cardiovascular morphogenesis. We then discuss their implications in the field of cardiovascular tissue engineering.
Collapse
Affiliation(s)
- Anne-Laure Duchemin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Helene Vignes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France.
| | - Renee Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
139
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
140
|
Biomechanical signaling within the developing zebrafish heart attunes endocardial growth to myocardial chamber dimensions. Nat Commun 2019; 10:4113. [PMID: 31511517 PMCID: PMC6739419 DOI: 10.1038/s41467-019-12068-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Intra-organ communication guides morphogenetic processes that are essential for an organ to carry out complex physiological functions. In the heart, the growth of the myocardium is tightly coupled to that of the endocardium, a specialized endothelial tissue that lines its interior. Several molecular pathways have been implicated in the communication between these tissues including secreted factors, components of the extracellular matrix, or proteins involved in cell-cell communication. Yet, it is unknown how the growth of the endocardium is coordinated with that of the myocardium. Here, we show that an increased expansion of the myocardial atrial chamber volume generates higher junctional forces within endocardial cells. This leads to biomechanical signaling involving VE-cadherin, triggering nuclear localization of the Hippo pathway transcriptional regulator Yap1 and endocardial proliferation. Our work suggests that the growth of the endocardium results from myocardial chamber volume expansion and ends when the tension on the tissue is relaxed. It is unknown how endocardium growth is coordinated with that of the myocardium in the zebrafish. Here, the authors show that myocardial chamber volume expansion causes increased endocardial tissue tension, which in turn triggers Hippo signaling-mediated proliferation within the endocardium.
Collapse
|
141
|
Yang Y, Cha B, Motawe ZY, Srinivasan RS, Scallan JP. VE-Cadherin Is Required for Lymphatic Valve Formation and Maintenance. Cell Rep 2019; 28:2397-2412.e4. [PMID: 31461654 PMCID: PMC6743082 DOI: 10.1016/j.celrep.2019.07.072] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/18/2019] [Accepted: 07/19/2019] [Indexed: 11/16/2022] Open
Abstract
The lymphatic vasculature requires intraluminal valves to maintain forward lymph flow. Lymphatic valves form and are constantly maintained by oscillatory fluid flow throughout life, yet the earliest steps of how lymphatic endothelial cells are able to respond to fluid shear stress remain unknown. Here, we show that the adherens junction protein VE-cadherin is required for the upregulation of valve-specific transcription factors. Conditional deletion of VE-cadherin in vivo prevented valve formation in the embryo and caused postnatal regression of nearly all lymphatic valves in multiple tissues. Since VE-cadherin is known to signal through β-catenin and the VEGFR/AKT pathway, each pathway was probed. Expression of a constitutively active β-catenin mutant or direct pharmacologic activation of AKT in vivo significantly rescued valve regression in the VE-cadherin-deficient lymphatic vessels. In conclusion, VE-cadherin-dependent signaling is required for lymphatic valve formation and maintenance and therapies to augment downstream pathways hold potential to treat lymphedema in patients.
Collapse
Affiliation(s)
- Ying Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Zeinab Y Motawe
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
142
|
Naicker T, Phoswa WN, Onyangunga OA, Gathiram P, Moodley J. Angiogenesis, Lymphangiogenesis, and the Immune Response in South African Preeclamptic Women Receiving HAART. Int J Mol Sci 2019; 20:ijms20153728. [PMID: 31366152 PMCID: PMC6696390 DOI: 10.3390/ijms20153728] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Purpose of the review: This review highlights the role of angiogenesis, lymphangiogenesis, and immune markers in human immunodeficiency virus (HIV)-associated preeclamptic (PE) pregnancies in an attempt to unravel the mysteries underlying the duality of both conditions in South Africa. Recent findings: Studies demonstrate that HIV-infected pregnant women develop PE at a lower frequency than uninfected women. In contrast, women receiving highly active anti-retroviral therapy (HAART) are more inclined to develop PE, stemming from an imbalance of angiogenesis, lymphangiogenesis, and immune response. Summary: In view of the paradoxical effect of HIV infection on PE development, this study examines angiogenesis, lymphangiogenesis, and immune markers in the highly HIV endemic area of KwaZulu-Natal. We believe that HAART re-constitutes the immune response in PE, thereby predisposing women to PE development. This susceptibility is due to an imbalance in the angiogenic/lymphangiogenic/immune response as compared to normotensive pregnant women. Further large-scale studies are urgently required to investigate the effect of the duration of HAART on PE development.
Collapse
Affiliation(s)
- Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4013, South Africa.
| | - Wendy N Phoswa
- Discipline of Obstetrics and Gynecology, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4013, South Africa.
| | - Onankoy A Onyangunga
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Premjith Gathiram
- Women's Health and HIV Research Group. Department of Obstetrics and Gynecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Jagidesa Moodley
- Women's Health and HIV Research Group. Department of Obstetrics and Gynecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4013, South Africa
| |
Collapse
|
143
|
Zanotelli MR, Reinhart-King CA. Mechanical Forces in Tumor Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1092:91-112. [PMID: 30368750 PMCID: PMC6986816 DOI: 10.1007/978-3-319-95294-9_6] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A defining hallmark of cancer and cancer development is upregulated angiogenesis. The vasculature formed in tumors is structurally abnormal, not organized in the conventional hierarchical arrangement, and more permeable than normal vasculature. These features contribute to leaky, tortuous, and dilated blood vessels, which act to create heterogeneous blood flow, compression of vessels, and elevated interstitial fluid pressure. As such, abnormalities in the tumor vasculature not only affect the delivery of nutrients and oxygen to the tumor, but also contribute to creating an abnormal tumor microenvironment that further promotes tumorigenesis. The role of chemical signaling events in mediating tumor angiogenesis has been well researched; however, the relative contribution of physical cues and mechanical regulation of tumor angiogenesis is less understood. Growing research indicates that the physical microenvironment plays a significant role in tumor progression and promoting abnormal tumor vasculature. Here, we review how mechanical cues found in the tumor microenvironment promote aberrant tumor angiogenesis. Specifically, we discuss the influence of matrix stiffness and mechanical stresses in tumor tissue on tumor vasculature, as well as the mechanosensory pathways utilized by endothelial cells to respond to the physical cues found in the tumor microenvironment. We also discuss the impact of the resulting aberrant tumor vasculature on tumor progression and therapeutic treatment.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
144
|
Regulatory mechanisms of Robo4 and their effects on angiogenesis. Biosci Rep 2019; 39:BSR20190513. [PMID: 31160487 PMCID: PMC6620384 DOI: 10.1042/bsr20190513] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
Roundabout4 (Robo4) is a transmembrane receptor that belongs to the Roundabout (Robo) family of axon guidance molecules. Robo4 is an endothelial-specific receptor that participates in endothelial cell migration, proliferation, and angiogenesis and the maintenance of vasculature homeostasis. The purpose of this review is to summarize and analyze three main mechanisms related to the expression and function of Robo4 during developmental and pathological angiogenesis. In this review, static shear stress and the binding of transcription factors such as E26 transformation-specific variant 2 (ETV2) and Slit3 induce Robo4 expression and activate Robo4 during tissue and organ development. Robo4 interacts with Slit2 or UNC5B to maintain vascular integrity, while a disturbed flow and the expression of transcription factors in inflammatory or neoplastic environments alter Robo4 expression levels, although these changes have uncertain functions. Based on the mechanisms described above, we discuss the aberrant expression of Robo4 in angiogenesis-related diseases and propose antiangiogenic therapies targeting the Robo4 signaling pathway for the treatment of ocular neovascularization lesions and tumors. Finally, although many problems related to Robo4 signaling pathways remain to be resolved, Robo4 is a promising and potentially valuable therapeutic target for treating pathological angiogenesis and developmental defects in angiogenesis.
Collapse
|
145
|
Zhong W, Yang W, Qin Y, Gu W, Xue Y, Tang Y, Xu H, Wang H, Zhang C, Wang C, Sun B, Liu Y, Liu H, Zhou H, Chen S, Sun T, Yang C. 6-Gingerol stabilized the p-VEGFR2/VE-cadherin/β-catenin/actin complex promotes microvessel normalization and suppresses tumor progression. J Exp Clin Cancer Res 2019; 38:285. [PMID: 31266540 PMCID: PMC6604152 DOI: 10.1186/s13046-019-1291-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Anti-angiogenic therapies demonstrate anti-tumor effects by decreasing blood supply to tumors and inhibiting tumor growth. However, anti-angiogenic therapy may leads to changes in tumor microenvironment and increased invasiveness of tumor cells, which in turn promotes distant metastasis and increased drug resistance. METHODS The CO-IP assays, N-STORM and cytoskeleton analysis were used to confirm the mechanism that p-VEGFR2/VE-cadherin/β-catenin/actin complex regulates vascular remodeling and improves the tumor microenvironment. 6-gingerol (6G), the major bioactive component in ginger, stabilized this complex by enhancing the binding of VEGFa to VEGFR2 with non-pathway dependent. Biacore, pull down and molecular docking were employed to confirm the interaction between 6G and VEGFR2 and enhancement of VEGFa binding to VEGFR2. RESULTS Here, we report that microvascular structural entropy (MSE) may be a prognostic factor in several tumor types and have potential as a biomarker in the clinic. 6G regulates the structural organization of the microvascular bed to decrease MSE via the p-VEGFR2/VE-cadherin/β-catenin/actin complex and inhibit tumor progression. 6G promotes the normalization of tumor vessels, improves the tumor microenvironment and decreases MSE, facilitating the delivery of chemotherapeutic agents into the tumor core and thereby reducing tumor growth and metastasis. CONCLUSIONS This study demonstrated the importance of vascular normalization in tumor therapy and elucidated the mechanism of action of ginger, a medicinal compound that has been used in China since ancient times.
Collapse
Affiliation(s)
- Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, 300041 China
| | - Wendong Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Yuan Qin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Wenguang Gu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
| | - Yinyin Xue
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Yuanhao Tang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Hengwei Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Hongzhi Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Chao Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Changhua Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
| | - Bo Sun
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Yanrong Liu
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Huijuan Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Shuang Chen
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350 China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300450 China
| |
Collapse
|
146
|
Babendreyer A, Molls L, Simons IM, Dreymueller D, Biller K, Jahr H, Denecke B, Boon RA, Bette S, Schnakenberg U, Ludwig A. The metalloproteinase ADAM15 is upregulated by shear stress and promotes survival of endothelial cells. J Mol Cell Cardiol 2019; 134:51-61. [PMID: 31271758 DOI: 10.1016/j.yjmcc.2019.06.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/18/2019] [Accepted: 06/28/2019] [Indexed: 02/08/2023]
Abstract
Reduced shear stress resulting from disturbed blood flow can impair endothelial integrity and drive the development of vascular inflammatory lesions. Metalloproteinases of the ADAM family have been implicated in the regulation of cell survival and inflammatory responses. Here we investigate the mechanism and function of ADAM15 upregulation in primary flow cultured endothelial cells. Transcriptomic analysis indicated that within the ADAM family ADAM15 mRNA is most prominently upregulated (4-fold) when endothelial cells are exposed to physiologic shear stress. This induction was confirmed in venous, arterial and microvascular endothelial cells and is associated with increased presence of ADAM15 protein in the cell lysates (5.6-fold) and on the surface (3.1-fold). The ADAM15 promoter contains several consensus sites for the transcription factor KLF2 which is also upregulated by shear stress. Induction of endothelial KLF2 by simvastatin treatment is associated with ADAM15 upregulation (1.8-fold) which is suppressed by counteracting simvastatin with geranylgeranyl pyrophosphate. KLF2 overexpression promotes ADAM15 expression (2.1-fold) under static conditions whereas KLF2 siRNA knockdown prevents ADAM15 induction by shear stress. Functionally, ADAM15 promotes survival of endothelial cells challenged by growth factor depletion or TNF stimulation as shown by ADAM15 shRNA knockdown (1.6-fold). Exposure to shear stress increases endothelial survival while additional knockdown of ADAM15 reduces survival (6.7-fold) under flow conditions. Thus, physiologic shear stress resulting from laminar flow promotes KLF2 induced ADAM15 expression which contributes to endothelial survival. The absence of ADAM15 at low shear stress or static conditions may therefore lead to increased endothelial damage and promote vascular inflammation.
Collapse
Affiliation(s)
- Aaron Babendreyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany.
| | - Lisa Molls
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Indra M Simons
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Kristina Biller
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Holger Jahr
- Institute of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany; Department of Orthopaedic Surgery, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Sebastian Bette
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - Uwe Schnakenberg
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
147
|
Iring A, Jin YJ, Albarrán-Juárez J, Siragusa M, Wang S, Dancs PT, Nakayama A, Tonack S, Chen M, Künne C, Sokol AM, Günther S, Martínez A, Fleming I, Wettschureck N, Graumann J, Weinstein LS, Offermanns S. Shear stress-induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J Clin Invest 2019; 129:2775-2791. [PMID: 31205027 DOI: 10.1172/jci123825] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 05/01/2019] [Indexed: 12/22/2022] Open
Abstract
Hypertension is a primary risk factor for cardiovascular diseases including myocardial infarction and stroke. Major determinants of blood pressure are vasodilatory factors such as nitric oxide (NO) released from the endothelium under the influence of fluid shear stress exerted by the flowing blood. Several endothelial signaling processes mediating fluid shear stress-induced formation and release of vasodilatory factors have been described. It is, however, still poorly understood how fluid shear stress induces these endothelial responses. Here we show that the endothelial mechanosensitive cation channel PIEZO1 mediated fluid shear stress-induced release of adrenomedullin, which in turn activated its Gs-coupled receptor. The subsequent increase in cAMP levels promoted the phosphorylation of endothelial NO synthase (eNOS) at serine 633 through protein kinase A (PKA), leading to the activation of the enzyme. This Gs/PKA-mediated pathway synergized with the AKT-mediated pathways leading to eNOS phosphorylation at serine 1177. Mice with endothelium-specific deficiency of adrenomedullin, the adrenomedullin receptor, or Gαs showed reduced flow-induced eNOS activation and vasodilation and developed hypertension. Our data identify fluid shear stress-induced PIEZO1 activation as a central regulator of endothelial adrenomedullin release and establish the adrenomedullin receptor and subsequent Gs-mediated formation of cAMP as a critical endothelial mechanosignaling pathway regulating basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Andras Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julián Albarrán-Juárez
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - Péter T Dancs
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Akiko Nakayama
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Anna M Sokol
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| | - Johannes Graumann
- German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
148
|
Boriushkin E, Fancher IS, Levitan I. Shear-Stress Sensitive Inwardly-Rectifying K + Channels Regulate Developmental Retinal Angiogenesis by Vessel Regression. Cell Physiol Biochem 2019; 52:1569-1583. [PMID: 31145841 PMCID: PMC7063968 DOI: 10.33594/000000109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIMS Shear stress plays major roles in developmental angiogenesis, particularly in blood vessel remodeling and maturation but little is known about the shear stress sensors involved in this process. Our recent study identified endothelial Kir2.1 channels as major contributors to flow-induced vasodilation, a hallmark of the endothelial flow response. The goal of this study is to establish the role of Kir2.1 in the regulation of retinal angiogenesis. METHODS The retina of newly born Kir2.1+/- mice were used to investigate the sprouting angiogenesis and remodeling of newly formed branched vessels. The structure, blood density and mural cell coverage have been evaluated by immunohistochemistry of the whole-mount retina. Endothelial cell alignment was assessed using CD31 staining. The experiments with flow-induced vasodilation were used to study the cerebrovascular response to flow. RESULTS Using Kir2.1-deficient mice, we show that the retinas of Kir2.1+/- mice have higher vessel density, increased lengths and increased number of the branching points, as compared to WT littermates. In contrast, the coverage by αSMA is decreased in Kir2.1+/- mice while pericyte coverage does not change. Furthermore, to determine whether deficiency of Kir2.1 affects vessel pruning, we discriminated between intact and degraded vessels or "empty matrix sleeves" and found a significant reduction in the number of empty sleeves on the peripheral part of the retina or "angiogenic front" in Kir2.1+/- mice. We also show that Kir2.1 deficiency results in decreased endothelial alignment in retinal endothelium and impaired flow-induced vasodilation of cerebral arteries, verifying the involvement of Kir2.1 in shear-stress sensing in retina and cerebral circulation. CONCLUSION This study shows that shear-stress sensitive Kir2.1 channels play an important role in pruning of excess vessels and vascular remodeling during retinal angiogenesis. We propose that Kir2.1 mediates the effect of shear stress on vessel maturation.
Collapse
Affiliation(s)
| | - Ibra S Fancher
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
149
|
LeBlanc ME, Saez-Torres KL, Cano I, Hu Z, Saint-Geniez M, Ng YS, D'Amore PA. Glycocalyx regulation of vascular endothelial growth factor receptor 2 activity. FASEB J 2019; 33:9362-9373. [PMID: 31141406 DOI: 10.1096/fj.201900011r] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have previously shown that knockdown of endomucin (EMCN), an integral membrane glycocalyx glycoprotein, prevents VEGF-induced proliferation, migration, and tube formation in vitro and angiogenesis in vivo. In the endothelium, VEGF mediates most of its angiogenic effects through VEGF receptor 2 (VEGFR2). To understand the role of EMCN, we examined the effect of EMCN depletion on VEGFR2 endocytosis and activation. Results showed that although VEGF stimulation promoted VEGFR2 internalization in control endothelial cells (ECs), loss of EMCN prevented VEGFR2 endocytosis. Cell surface analysis revealed a decrease in VEGFR2 following VEGF stimulation in control but not siRNA directed against EMCN-transfected ECs. EMCN depletion resulted in heightened phosphorylation following VEGF stimulation with an increase in total VEGFR2 protein. These results indicate that EMCN modulates VEGFR2 endocytosis and activity and point to EMCN as a potential therapeutic target.-LeBlanc, M. E., Saez-Torres, K. L., Cano, I., Hu, Z., Saint-Geniez, M., Ng, Y.-S., D'Amore, P. A. Glycocalyx regulation of vascular endothelial growth factor receptor 2 activity.
Collapse
Affiliation(s)
- Michelle E LeBlanc
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kahira L Saez-Torres
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Issahy Cano
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yin-Shan Ng
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
150
|
Man HSJ, Marsden PA. LncRNAs and epigenetic regulation of vascular endothelium: genome positioning system and regulators of chromatin modifiers. Curr Opin Pharmacol 2019; 45:72-80. [PMID: 31125866 DOI: 10.1016/j.coph.2019.04.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/15/2019] [Indexed: 12/31/2022]
Abstract
Epigenetic mechanisms regulate the cell type-specific expression of endothelial-enriched genes. A major question has been how chromatin modifiers without inherent sequence specificity can be targeted to genomic coordinates. Recently, long noncoding RNAs (lncRNAs) have emerged as candidates for specifying genomic positioning for chromatin modifiers. However, lncRNAs function by a number of mechanisms in both the nucleus and the cytoplasm. Recent studies indicate the existence of endothelial-enriched lncRNAs. This review discusses lncRNA regulation in endothelial cells with a focus on four recently described nuclear-enriched lncRNAs: MANTIS, LEENE, STEEL, and GATA6-AS. This emerging work on these lncRNAs contributes to our understanding of epigenetic regulation in the vascular endothelium with links to important themes in endothelial biology, including angiogenesis and shear stress.
Collapse
Affiliation(s)
- Hon-Sum Jeffrey Man
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Philip A Marsden
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|