1
|
Roth M, Carlsson R, Buizza C, Enström A, Paul G. Pericyte response to ischemic stroke precedes endothelial cell death and blood-brain barrier breakdown. J Cereb Blood Flow Metab 2025; 45:617-629. [PMID: 39053491 PMCID: PMC11571979 DOI: 10.1177/0271678x241261946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 07/27/2024]
Abstract
Stroke is one of the leading causes of death and disability, yet the cellular response to the ischemic insult is poorly understood limiting therapeutic options. Brain pericytes are crucial for maintaining blood-brain barrier (BBB) integrity and are known to be one of the first responders to ischemic stroke. The exact timeline of cellular events after stroke, however, remains elusive. Using the permanent middle cerebral artery occlusion stroke model, we established a detailed timeline of microvascular events after experimental stroke. Our results show that pericytes respond already within 1 hour after the ischemic insult. We find that approximately 30% of the pericyte population dies as early as 1 hour after stroke, while ca 50% express markers that indicate activation. A decrease of endothelial tight junctions, signs of endothelial cell death and reduction in blood vessel length are only detected at time points after the initial pericyte response. Consistently, markers of BBB leakage are observed several hours after pericyte cell death and/or vascular detachment. Our results suggest that the pericyte response to stroke occurs early and precedes both the endothelial response and the BBB breakdown. This highlights pericytes as an important target cell type to develop new diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Michaela Roth
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Carolina Buizza
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
| |
Collapse
|
2
|
Sheibani N, Sang Y, Wang S, Sorenson CM. Bax expression impacts postnatal retinal vascular development and hyperoxia sensitivity. Exp Eye Res 2024; 248:110107. [PMID: 39307450 PMCID: PMC11532006 DOI: 10.1016/j.exer.2024.110107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
Apoptosis plays prominent roles during organ development, maturation and homeostasis. In the retina, Bcl-2 family members function through the intrinsic cell death pathway with vital roles during vascular development and hyperoxia-mediated vessel obliteration during oxygen induced ischemic retinopathy (OIR). Bim, a BH3 only protein Bcl-2 family member, binds and activates Bax and/or Bak to facilitate apoptosis. In some systems deletion of both Bax and Bak are required to prevent cell loss, such as regression of ocular hyaloid vasculature. We previously showed Bim expression significantly impacts normal retinal vascular development and sensitivity to hyperoxia. Mice deficient in Bim (Bim-/-) show increased retinal vascular density and are protected from hyperoxia mediated vessel obliteration. Since Bim activates Bax, here we determined the impact lack of Bax expression has on these processes. Compared to Bax+/+ mice, retinas from Bax-/- mice had significantly increased numbers of retinal endothelial cells and pericytes. We also demonstrated that hyperoxia-mediated vessel obliteration during OIR was significantly decreased in the absence of Bax. Although the increased endothelial cell numbers were comparable to that of Bim-/- mice, the increased numbers of pericytes were not to the extent noted in Bim-/- mice. These changes were supported by partial protection of retinal vessels from hyperoxia in Bax-/- mice compared to that noted in Bim-/- mice. Thus, Bim-Bax driven pathway is sufficient to remove excess endothelial cells but not pericytes during postnatal retinal vascularization and hyperoxia-mediated vessel obliteration. Thus, additional Bim-mediated pathway(s) are required for removal of pericytes and hyperoxia-mediated vessel obliteration.
Collapse
Affiliation(s)
- Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Yanzhi Sang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christine M Sorenson
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
3
|
Shen X, Kong F, Wen J, Wang X, Huang C. The role of inflammation in central serous chorioretinopathy: From mechanisms to therapeutic prospects. Front Pharmacol 2024; 15:1200492. [PMID: 38835666 PMCID: PMC11148560 DOI: 10.3389/fphar.2024.1200492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Central serous chorioretinopathy (CSC) is a leading cause of permanent vision loss, ranking fourth among macular diseases, trailing only age-related macular degeneration, diabetic retinopathy, and retinal vein obstruction. While mounting evidence implicates inflammation as a pivotal factor in the onset and advancement of CSC, the specific pathophysiological process and molecular mechanisms underlying inflammation remain incompletely understood. A complex network of cytokines, chemokines, and adhesion molecules interplay to trigger inflammatory and pathological cascades, highlighting the need for a comprehensive comprehension of the inflammation-related mechanisms behind CSC progression. In this piece, we examine the existing comprehension of CSC's pathology and pathogenesis. Additionally, we present an overview of the mechanisms underlying the onset and progression of CSC inflammation, followed by a thorough analysis and discussion of the potential of targeted inflammatory intervention for both preventing and treating CSC.
Collapse
Affiliation(s)
- Xiao Shen
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Fanhua Kong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Jing Wen
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Xiao Wang
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Chunlian Huang
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
4
|
Zhao YQ, Ren YF, Li BB, Wei C, Yu B. The mysterious association between adiponectin and endometriosis. Front Pharmacol 2024; 15:1396616. [PMID: 38813109 PMCID: PMC11133721 DOI: 10.3389/fphar.2024.1396616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Adiponectin is a pleiotropic cytokine predominantly derived from adipose tissue. In addition to its role in regulating energy metabolism, adiponectin may also be related to estrogen-dependent diseases, and many studies have confirmed its involvement in mediating diverse biological processes, including apoptosis, autophagy, inflammation, angiogenesis, and fibrosis, all of which are related to the pathogenesis of endometriosis. Although many researchers have reported low levels of adiponectin in patients with endometriosis and suggested that it may serve as a protective factor against the development of the disease. Therefore, the purpose of this review was to provide an up-to-date summary of the roles of adiponectin and its downstream cytokines and signaling pathways in the aforementioned biological processes. Further systematic studies on the molecular and cellular mechanisms of action of adiponectin may provide novel insights into the pathophysiology of endometriosis as well as potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Bing-Bing Li
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, Shandong Province, China
| | | | | |
Collapse
|
5
|
He J, Blazeski A, Nilanthi U, Menéndez J, Pirani SC, Levic DS, Bagnat M, Singh MK, Raya JG, García-Cardeña G, Torres-Vázquez J. Plxnd1-mediated mechanosensing of blood flow controls the caliber of the Dorsal Aorta via the transcription factor Klf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.576555. [PMID: 38328196 PMCID: PMC10849625 DOI: 10.1101/2024.01.24.576555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The cardiovascular system generates and responds to mechanical forces. The heartbeat pumps blood through a network of vascular tubes, which adjust their caliber in response to the hemodynamic environment. However, how endothelial cells in the developing vascular system integrate inputs from circulatory forces into signaling pathways to define vessel caliber is poorly understood. Using vertebrate embryos and in vitro-assembled microvascular networks of human endothelial cells as models, flow and genetic manipulations, and custom software, we reveal that Plexin-D1, an endothelial Semaphorin receptor critical for angiogenic guidance, employs its mechanosensing activity to serve as a crucial positive regulator of the Dorsal Aorta's (DA) caliber. We also uncover that the flow-responsive transcription factor KLF2 acts as a paramount mechanosensitive effector of Plexin-D1 that enlarges endothelial cells to widen the vessel. These findings illuminate the molecular and cellular mechanisms orchestrating the interplay between cardiovascular development and hemodynamic forces.
Collapse
Affiliation(s)
- Jia He
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Uthayanan Nilanthi
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
| | - Javier Menéndez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Samuel C. Pirani
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel S. Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Manvendra K. Singh
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609
| | - José G Raya
- Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesús Torres-Vázquez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
6
|
Shi Z, Yao C, Shui Y, Li S, Yan H. Research progress on the mechanism of angiogenesis in wound repair and regeneration. Front Physiol 2023; 14:1284981. [PMID: 38089479 PMCID: PMC10711283 DOI: 10.3389/fphys.2023.1284981] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 12/10/2024] Open
Abstract
Poor wound healing and pathological healing have been pressing issues in recent years, as they impact human quality of life and pose risks of long-term complications. The study of neovascularization has emerged as a prominent research focus to address these problems. During the process of repair and regeneration, the establishment of a new vascular system is an indispensable stage for complete healing. It provides favorable conditions for nutrient delivery, oxygen supply, and creates an inflammatory environment. Moreover, it is a key manifestation of the proliferative phase of wound healing, bridging the inflammatory and remodeling phases. These three stages are closely interconnected and inseparable. This paper comprehensively integrates the regulatory mechanisms of new blood vessel formation in wound healing, focusing on the proliferation and migration of endothelial cells and the release of angiogenesis-related factors under different healing outcomes. Additionally, the hidden link between the inflammatory environment and angiogenesis in wound healing is explored.
Collapse
Affiliation(s)
- Zhuojun Shi
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Chong Yao
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Yujie Shui
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Site Li
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Hong Yan
- Laboratory of Plastic Surgery, Department of Plastic Surgery and Reconstruction, Second Hospital of West China, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Wen L, Yan W, Zhu L, Tang C, Wang G. The role of blood flow in vessel remodeling and its regulatory mechanism during developmental angiogenesis. Cell Mol Life Sci 2023; 80:162. [PMID: 37221410 PMCID: PMC11072276 DOI: 10.1007/s00018-023-04801-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/06/2023] [Accepted: 05/06/2023] [Indexed: 05/25/2023]
Abstract
Vessel remodeling is essential for a functional and mature vascular network. According to the difference in endothelial cell (EC) behavior, we classified vessel remodeling into vessel pruning, vessel regression and vessel fusion. Vessel remodeling has been proven in various organs and species, such as the brain vasculature, subintestinal veins (SIVs), and caudal vein (CV) in zebrafish and yolk sac vessels, retina, and hyaloid vessels in mice. ECs and periendothelial cells (such as pericytes and astrocytes) contribute to vessel remodeling. EC junction remodeling and actin cytoskeleton dynamic rearrangement are indispensable for vessel pruning. More importantly, blood flow has a vital role in vessel remodeling. In recent studies, several mechanosensors, such as integrins, platelet endothelial cell adhesion molecule-1 (PECAM-1)/vascular endothelial cell (VE-cadherin)/vascular endothelial growth factor receptor 2 (VEGFR2) complex, and notch1, have been shown to contribute to mechanotransduction and vessel remodeling. In this review, we highlight the current knowledge of vessel remodeling in mouse and zebrafish models. We further underline the contribution of cellular behavior and periendothelial cells to vessel remodeling. Finally, we discuss the mechanosensory complex in ECs and the molecular mechanisms responsible for vessel remodeling.
Collapse
Affiliation(s)
- Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wenhua Yan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Zhu
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, 215123, China
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, 215123, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- JinFeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
8
|
Whitehead L, Watson EC, Grant ZL. Analyzing Vessel Regression and Endothelial Apoptosis as a Component of Angiogenic Vessel Remodeling. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:29-39. [PMID: 35099726 DOI: 10.1007/978-1-0716-2059-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Angiogenic vessel remodeling is a critical step in establishing a hierarchical vessel network. Vessel networks rapidly expand through angiogenesis in response to pro-angiogenic factors. This leads to an initially dense vessel network that requires selective regression of vessel branches to establish a hierarchical conduit for blood flow, a process known as pruning. This involves migration of endothelial cells from low-flow vessels to adjacent high-flow vessels and generally occurs independently of cell death. Vessels may also regress in response to other stimuli, including reduced metabolic demand, redundancy, and pathological stimuli. In these contexts, widespread vessel regression typically occurs and involves loss of endothelial cells by apoptotic cell death. Thus, vessel remodeling occurs via both apoptosis independent and dependent vessel regression. In this chapter, we outline a semi-automated method for quantifying vessel regression using the neonatal model of angiogenesis. We further provide instruction on analyzing endothelial apoptosis in this model.
Collapse
Affiliation(s)
- Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Emma C Watson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Faculty of Medicine, Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Zoe L Grant
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
- Gladstone Institutes, San Francisco, CA, USA.
| |
Collapse
|
9
|
Takino JI, Miyazaki S, Nagamine K, Hori T. The Role of RASGRP2 in Vascular Endothelial Cells-A Mini Review. Int J Mol Sci 2021; 22:ijms222011129. [PMID: 34681791 PMCID: PMC8537898 DOI: 10.3390/ijms222011129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
RAS guanyl nucleotide-releasing proteins (RASGRPs) are important proteins that act as guanine nucleotide exchange factors, which activate small GTPases and function as molecular switches for intracellular signals. The RASGRP family is composed of RASGRP1-4 proteins and activates the small GTPases, RAS and RAP. Among them, RASGRP2 has different characteristics from other RASGRPs in that it targets small GTPases and its localizations are different. Many studies related to RASGRP2 have been reported in cells of the blood cell lineage. Furthermore, RASGRP2 has also been reported to be associated with Huntington's disease, tumors, and rheumatoid arthritis. In addition, we also recently reported RASGRP2 expression in vascular endothelial cells, and clarified the involvement of xenopus Rasgrp2 in the vasculogenesis process and multiple signaling pathways of RASGRP2 in human vascular endothelial cells with stable expression of RASGRP2. Therefore, this article outlines the existing knowledge of RASGRP2 and focuses on its expression and role in vascular endothelial cells, and suggests that RASGRP2 functions as a protective factor for maintaining healthy blood vessels.
Collapse
Affiliation(s)
- Jun-ichi Takino
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
- Correspondence: ; Tel.: +81-823-73-8584
| | - Shouhei Miyazaki
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
| | - Kentaro Nagamine
- Faculty of Health Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan;
| | - Takamitsu Hori
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
| |
Collapse
|
10
|
Ben Dhaou C, Mandi K, Frye M, Acheampong A, Radi A, De Becker B, Antoine M, Baeyens N, Wittamer V, Parmentier M. Chemerin regulates normal angiogenesis and hypoxia-driven neovascularization. Angiogenesis 2021; 25:159-179. [PMID: 34524600 PMCID: PMC9054887 DOI: 10.1007/s10456-021-09818-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/05/2021] [Indexed: 02/01/2023]
Abstract
Chemerin is a multifunctional protein initially characterized in our laboratory as a chemoattractant factor for leukocyte populations. Its main functional receptor is CMKLR1. We identified previously chemerin as an anti-tumoral factor inhibiting the vascularization of tumor grafts. We show here that overexpression of bioactive chemerin in mice results in a reduction of the density of the retinal vascular network during its development and in adults. Chemerin did not affect vascular sprouting during the post-natal development of the network, but rather promoted endothelial cell apoptosis and vessel pruning. This phenotype was reversed to normal in CMKLR1-deficient mice, demonstrating the role of this receptor. Chemerin inhibited also neoangiogenesis in a model of pathological proliferative retinopathy, and in response to hind-limb ischemia. Mechanistically, PTEN and FOXO1 antagonists could almost completely restore the density of the retinal vasculature, suggesting the involvement of the PI3-kinase/AKT pathway in the chemerin-induced vessel regression process.
Collapse
Affiliation(s)
- Cyrine Ben Dhaou
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium.,Physiologie de la Reproduction et des Comportements, University of Tours, INRA Val-de-Loire UMR-85, CNRS UMR-1247, Tours, France
| | - Kamel Mandi
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium
| | - Mickaël Frye
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium
| | - Angela Acheampong
- Cardiology Department, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Ayoub Radi
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium
| | - Benjamin De Becker
- Cardiology Department, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Mathieu Antoine
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium
| | - Nicolas Baeyens
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium
| | - Valérie Wittamer
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium
| | - Marc Parmentier
- WELBIO and I.R.I.B.H.M, Université Libre de Bruxelles, Campus Erasme, 808 route de Lennik, B-1070, Brussels, Belgium.
| |
Collapse
|
11
|
A ligand-insensitive UNC5B splicing isoform regulates angiogenesis by promoting apoptosis. Nat Commun 2021; 12:4872. [PMID: 34381052 PMCID: PMC8358048 DOI: 10.1038/s41467-021-24998-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The Netrin-1 receptor UNC5B is an axon guidance regulator that is also expressed in endothelial cells (ECs), where it finely controls developmental and tumor angiogenesis. In the absence of Netrin-1, UNC5B induces apoptosis that is blocked upon Netrin-1 binding. Here, we identify an UNC5B splicing isoform (called UNC5B-Δ8) expressed exclusively by ECs and generated through exon skipping by NOVA2, an alternative splicing factor regulating vascular development. We show that UNC5B-Δ8 is a constitutively pro-apoptotic splicing isoform insensitive to Netrin-1 and required for specific blood vessel development in an apoptosis-dependent manner. Like NOVA2, UNC5B-Δ8 is aberrantly expressed in colon cancer vasculature where its expression correlates with tumor angiogenesis and poor patient outcome. Collectively, our data identify a mechanism controlling UNC5B’s necessary apoptotic function in ECs and suggest that the NOVA2/UNC5B circuit represents a post-transcriptional pathway regulating angiogenesis. UNC5B is a Netrin-1 receptor expressed in endothelial cells that in the absence of ligand induces apoptosis. Here the authors identify an UNC5B splicing isoform that is insensitive to the pro-survival ligand Netrin-1 and is required for apoptosis-dependent blood vessel development.
Collapse
|
12
|
Tisch N, Ruiz de Almodóvar C. Contribution of cell death signaling to blood vessel formation. Cell Mol Life Sci 2021; 78:3247-3264. [PMID: 33783563 PMCID: PMC8038986 DOI: 10.1007/s00018-020-03738-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
The formation of new blood vessels is driven by proliferation of endothelial cells (ECs), elongation of maturing vessel sprouts and ultimately vessel remodeling to create a hierarchically structured vascular system. Vessel regression is an essential process to remove redundant vessel branches in order to adapt the final vessel density to the demands of the surrounding tissue. How exactly vessel regression occurs and whether and to which extent cell death contributes to this process has been in the focus of several studies within the last decade. On top, recent findings challenge our simplistic view of the cell death signaling machinery as a sole executer of cellular demise, as emerging evidences suggest that some of the classic cell death regulators even promote blood vessel formation. This review summarizes our current knowledge on the role of the cell death signaling machinery with a focus on the apoptosis and necroptosis signaling pathways during blood vessel formation in development and pathology.
Collapse
Affiliation(s)
- Nathalie Tisch
- Department of Vascular Dysfunction, European Center for Angioscience (ECAS), Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carmen Ruiz de Almodóvar
- Department of Vascular Dysfunction, European Center for Angioscience (ECAS), Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
13
|
Grant ZL, Whitehead L, Wong VH, He Z, Yan RY, Miles AR, Benest AV, Bates DO, Prahst C, Bentley K, Bui BV, Symons RC, Coultas L. Blocking endothelial apoptosis revascularizes the retina in a model of ischemic retinopathy. J Clin Invest 2021; 130:4235-4251. [PMID: 32427589 PMCID: PMC7410052 DOI: 10.1172/jci127668] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
Aberrant, neovascular retinal blood vessel growth is a vision-threatening complication in ischemic retinal diseases. It is driven by retinal hypoxia frequently caused by capillary nonperfusion and endothelial cell (EC) loss. We investigated the role of EC apoptosis in this process using a mouse model of ischemic retinopathy, in which vessel closure and EC apoptosis cause capillary regression and retinal ischemia followed by neovascularization. Protecting ECs from apoptosis in this model did not prevent capillary closure or retinal ischemia. Nonetheless, it prevented the clearance of ECs from closed capillaries, delaying vessel regression and allowing ECs to persist in clusters throughout the ischemic zone. In response to hypoxia, these preserved ECs underwent a vessel sprouting response and rapidly reassembled into a functional vascular network. This alleviated retinal hypoxia, preventing subsequent pathogenic neovascularization. Vessel reassembly was not limited by VEGFA neutralization, suggesting it was not dependent on the excess VEGFA produced by the ischemic retina. Neutralization of ANG2 did not prevent vessel reassembly, but did impair subsequent angiogenic expansion of the reassembled vessels. Blockade of EC apoptosis may promote ischemic tissue revascularization by preserving ECs within ischemic tissue that retain the capacity to reassemble a functional network and rapidly restore blood supply.
Collapse
Affiliation(s)
- Zoe L Grant
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, and
| | - Lachlan Whitehead
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, and
| | - Vickie Hy Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Richard Y Yan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Abigail R Miles
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Andrew V Benest
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, School of Medicine, University of Nottingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Birmingham, United Kingdom
| | - David O Bates
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, School of Medicine, University of Nottingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Birmingham, United Kingdom
| | - Claudia Prahst
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Katie Bentley
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA.,Beijer Laboratory for Gene and Neuroscience Research, Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Robert Ca Symons
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia.,Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Department of Ophthalmology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Leigh Coultas
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, and
| |
Collapse
|
14
|
Ehling M, Celus W, Martín-Pérez R, Alba-Rovira R, Willox S, Ponti D, Cid MC, Jones EAV, Di Conza G, Mazzone M. B55α/PP2A Limits Endothelial Cell Apoptosis During Vascular Remodeling: A Complementary Approach To Disrupt Pathological Vessels? Circ Res 2020; 127:707-723. [PMID: 32527198 PMCID: PMC7616433 DOI: 10.1161/circresaha.119.316071] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 06/11/2020] [Indexed: 12/16/2022]
Abstract
RATIONALE How endothelial cells (ECs) migrate and form an immature vascular plexus has been extensively studied. Yet, mechanisms underlying vascular remodeling remain poorly established. A better understanding of these processes may lead to the design of novel therapeutic strategies complementary to current angiogenesis inhibitors. OBJECTIVE Starting from our previous observations that PP2A (protein phosphatase 2) regulates the HIF (hypoxia-inducible factor)/PHD-2 (prolyl hydroxylase 2)-constituted oxygen machinery, we hypothesized that this axis could play an important role during blood vessel formation, tissue perfusion, and oxygen restoration. METHODS AND RESULTS We show that the PP2A regulatory subunit B55α is at the crossroad between vessel pruning and vessel maturation. Blood vessels with high B55α counter cell stress conditions and thrive for stabilization and maturation. When B55α is inhibited, ECs cannot cope with cell stress and undergo apoptosis, leading to massive pruning of nascent blood vessels. Mechanistically, we found that the B55α/PP2A complex restrains PHD-2 activity, promoting EC survival in a HIF-dependent manner, and furthermore dephosphorylates p38, altogether protecting ECs against cell stress occurring, for example, during the onset of blood flow. In tumors, EC-specific B55α deficiency induces pruning of immature-like tumor blood vessels resulting in delayed tumor growth and metastasis, without affecting nonpathological vessels. Consistently, systemic administration of a pan-PP2A inhibitor disrupts vascular network formation and tumor progression in vivo without additional effects on B55α-deficient vessels. CONCLUSIONS Our data underline a unique role of the B55α/PP2A phosphatase complex in vessel remodeling and suggest the use of PP2A-inhibitors as potent antiangiogenic drugs targeting specifically nascent blood vessels with a mode-of-action complementary to VEGF-R (vascular endothelial growth factor receptor)-targeted therapies. Graphical Abstract: A graphical abstract is available for this article.
Collapse
Affiliation(s)
- Manuel Ehling
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
| | - Ward Celus
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
| | - Rosa Martín-Pérez
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
| | - Roser Alba-Rovira
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
- Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona (R.A.-R., M.C.C.)
| | - Sander Willox
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
| | - Donatella Ponti
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
- Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Latina (D.P.)
| | - Maria C Cid
- Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona (R.A.-R., M.C.C.)
| | | | - Giusy Di Conza
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
| | - Massimiliano Mazzone
- From the Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.)
- Laboratory of Tumor Inflammation and Angiogenesis, and Department of Oncology (M.E., W.C., R.M.-P., R.A.-R., S.W., D.P., G.D.C., M.M.), KU Leuven, Belgium
| |
Collapse
|
15
|
Ding R, Hase Y, Ameen-Ali KE, Ndung'u M, Stevenson W, Barsby J, Gourlay R, Akinyemi T, Akinyemi R, Uemura MT, Polvikoski T, Mukaetova-Ladinska E, Ihara M, Kalaria RN. Loss of capillary pericytes and the blood-brain barrier in white matter in poststroke and vascular dementias and Alzheimer's disease. Brain Pathol 2020; 30:1087-1101. [PMID: 32705757 PMCID: PMC8018063 DOI: 10.1111/bpa.12888] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/29/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023] Open
Abstract
White matter (WM) disease is associated with disruption of the gliovascular unit, which involves breach of the blood–brain barrier (BBB). We quantified pericytes as components of the gliovascular unit and assessed their status in vascular and other common dementias. Immunohistochemical and immunofluorescent methods were developed to assess the distribution and quantification of pericytes connected to the frontal lobe WM capillaries. Pericytes with a nucleus were identified by collagen 4 (COL4) and platelet‐derived growth factor receptor‐β (PDGFR‐β) antibodies with further verification using PDGFR‐β‐specific ELISA. We evaluated a total of 124 post‐mortem brains from subjects with post‐stroke dementia (PSD), vascular dementia (VaD), Alzheimer’s disease (AD), AD‐VaD (Mixed) and post‐stroke non‐demented (PSND) stroke survivors as well as normal aging controls. COL4 and PDGFR‐β reactive pericytes adopted the characteristic “crescent” or nodule‐like shapes around capillary walls. We estimated densities of pericyte somata to be 225 ±38 and 200 ±13 (SEM) per COL4 mm2 area or 2.0 ± 0.1 and 1.7 ± 0.1 per mm capillary length in young and older aging controls. Remarkably, WM pericytes were reduced by ~35%–45% in the frontal lobe of PSD, VaD, Mixed and AD subjects compared to PSND and controls subjects (P < 0.001). We also found pericyte numbers were correlated with PDGFR‐β reactivity in the WM. Our results first demonstrate a reliable method to quantify COL4‐positive pericytes and then, indicate that deep WM pericytes are decreased across different dementias including PSD, VaD, Mixed and AD. Our findings suggest that downregulation of pericytes is associated with the disruption of the BBB in the deep WM in several aging‐related dementias.
Collapse
Affiliation(s)
- Ren Ding
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - Yoshiki Hase
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - Kamar E Ameen-Ali
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - Michael Ndung'u
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - William Stevenson
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - Joseph Barsby
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - Ryan Gourlay
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | - Tolulope Akinyemi
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Rufus Akinyemi
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Maiko T Uemura
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tuomo Polvikoski
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| | | | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Raj N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne, UK
| |
Collapse
|
16
|
Ko VH, Yu LJ, Dao DT, Li X, Secor JD, Anez-Bustillos L, Cho BS, Pan A, Mitchell PD, Kishikawa H, Puder M. Roxadustat (FG-4592) accelerates pulmonary growth, development, and function in a compensatory lung growth model. Angiogenesis 2020; 23:637-649. [PMID: 32666268 DOI: 10.1007/s10456-020-09735-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Children with hypoplastic lung disease associated with congenital diaphragmatic hernia (CDH) continue to suffer significant morbidity and mortality secondary to progressive pulmonary disease. Current management of CDH is primarily supportive and mortality rates of the most severely affected children have remained unchanged in the last few decades. Previous work in our lab has demonstrated the importance of vascular endothelial growth factor (VEGF)-mediated angiogenesis in accelerating compensatory lung growth. In this study, we evaluated the potential for Roxadustat (FG-4592), a prolyl hydroxylase inhibitor known to increase endogenous VEGF, in accelerating compensatory lung growth. Treatment with Roxadustat increased lung volume, total lung capacity, alveolarization, and exercise tolerance compared to controls following left pneumonectomy. However, this effect was likely modulated not only by increased VEGF, but rather also by decreased pigment epithelium-derived factor (PEDF), an anti-angiogenic factor. Furthermore, this mechanism of action may be specific to Roxadustat. Vadadustat (AKB-6548), a structurally similar prolyl hydroxylase inhibitor, did not demonstrate accelerated compensatory lung growth or decreased PEDF expression following left pneumonectomy. Given that Roxadustat is already in Phase III clinical studies for the treatment of chronic kidney disease-associated anemia with minimal side effects, its use for the treatment of pulmonary hypoplasia could potentially proceed expeditiously.
Collapse
Affiliation(s)
- Victoria H Ko
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Lumeng J Yu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Duy T Dao
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Xiaoran Li
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Jordan D Secor
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Lorenzo Anez-Bustillos
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Bennet S Cho
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Amy Pan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Paul D Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Hiroko Kishikawa
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Mark Puder
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA. .,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Fonseca CG, Barbacena P, Franco CA. Endothelial cells on the move: dynamics in vascular morphogenesis and disease. VASCULAR BIOLOGY 2020; 2:H29-H43. [PMID: 32935077 PMCID: PMC7487603 DOI: 10.1530/vb-20-0007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
The vascular system is a hierarchically organized network of blood vessels that play crucial roles in embryogenesis, homeostasis and disease. Blood vessels are built by endothelial cells – the cells lining the interior of blood vessels – through a process named vascular morphogenesis. Endothelial cells react to different biomechanical signals in their environment by adjusting their behavior to: (1) invade, proliferate and fuse to form new vessels (angiogenesis); (2) remodel, regress and establish a hierarchy in the network (patterning); and (3) maintain network stability (quiescence). Each step involves the coordination of endothelial cell differentiation, proliferation, polarity, migration, rearrangements and shape changes to ensure network integrity and an efficient barrier between blood and tissues. In this review, we highlighted the relevance and the mechanisms involving endothelial cell migration during different steps of vascular morphogenesis. We further present evidence on how impaired endothelial cell dynamics can contribute to pathology.
Collapse
Affiliation(s)
- Catarina G Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Barbacena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Claudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
18
|
Tisch N, Freire-Valls A, Yerbes R, Paredes I, La Porta S, Wang X, Martín-Pérez R, Castro L, Wong WWL, Coultas L, Strilic B, Gröne HJ, Hielscher T, Mogler C, Adams RH, Heiduschka P, Claesson-Welsh L, Mazzone M, López-Rivas A, Schmidt T, Augustin HG, Ruiz de Almodovar C. Caspase-8 modulates physiological and pathological angiogenesis during retina development. J Clin Invest 2020; 129:5092-5107. [PMID: 31454332 DOI: 10.1172/jci122767] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/20/2019] [Indexed: 12/29/2022] Open
Abstract
During developmental angiogenesis, blood vessels grow and remodel to ultimately build a hierarchical vascular network. Whether, how, cell death signaling molecules contribute to blood vessel formation is still not well understood. Caspase-8 (Casp-8), a key protease in the extrinsic cell death-signaling pathway, regulates cell death via both apoptosis and necroptosis. Here, we show that expression of Casp-8 in endothelial cells (ECs) is required for proper postnatal retina angiogenesis. EC-specific Casp-8-KO pups (Casp-8ECKO) showed reduced retina angiogenesis, as the loss of Casp-8 reduced EC proliferation, sprouting, and migration independently of its cell death function. Instead, the loss of Casp-8 caused hyperactivation of p38 MAPK downstream of receptor-interacting serine/threonine protein kinase 3 (RIPK3) and destabilization of vascular endothelial cadherin (VE-cadherin) at EC junctions. In a mouse model of oxygen-induced retinopathy (OIR) resembling retinopathy of prematurity (ROP), loss of Casp-8 in ECs was beneficial, as pathological neovascularization was reduced in Casp-8ECKO pups. Taking these data together, we show that Casp-8 acts in a cell death-independent manner in ECs to regulate the formation of the retina vasculature and that Casp-8 in ECs is mechanistically involved in the pathophysiology of ROP.
Collapse
Affiliation(s)
- Nathalie Tisch
- Biochemistry Center.,European Center for Angioscience (ECAS).,Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, and
| | - Aida Freire-Valls
- Biochemistry Center.,Department of General, Visceral and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Rosario Yerbes
- Biochemistry Center.,Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Sevilla and Universidad Pablo de Olavide, Sevilla, Spain
| | - Isidora Paredes
- Biochemistry Center.,European Center for Angioscience (ECAS).,Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, and
| | - Silvia La Porta
- European Center for Angioscience (ECAS).,Division of Vascular Oncology and Metastasis, German Cancer Research Center, Heidelberg, Germany
| | | | - Rosa Martín-Pérez
- Lab of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (VIB), Leuven, Belgium.,Lab of Tumor Inflammation and Angiogenesis, Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | - Wendy Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Leigh Coultas
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine and
| | - Peter Heiduschka
- Research Laboratory, Department of Ophthalmology, University Medical Center, University of Münster, Münster, Germany
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Massimiliano Mazzone
- Lab of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (VIB), Leuven, Belgium.,Lab of Tumor Inflammation and Angiogenesis, Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Abelardo López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Sevilla and Universidad Pablo de Olavide, Sevilla, Spain.,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Hellmut G Augustin
- European Center for Angioscience (ECAS).,Division of Vascular Oncology and Metastasis, German Cancer Research Center, Heidelberg, Germany
| | - Carmen Ruiz de Almodovar
- Biochemistry Center.,European Center for Angioscience (ECAS).,Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, and
| |
Collapse
|
19
|
Jiao C, Adler K, Liu X, Sun W, Mullins RF, Sohn EH. Visualization of Mouse Choroidal and Retinal Vasculature Using Fluorescent Tomato Lectin Perfusion. Transl Vis Sci Technol 2020; 9:1. [PMID: 32509436 PMCID: PMC7255627 DOI: 10.1167/tvst.9.1.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose To develop a reliable and simplified method to assess choroid and retinal vasculature on whole mount and cross sections in mice using tomato lectin (TL; Lycopersicon esculentum). Methods Albino mice (n = 27) received 1 mg/mL of TL (conjugated to Dylight-594) intravascularly through the tail vein, jugular vein, or cardiac left ventricle. Whole mounts of the retina and choroid were evaluated using fluorescence microscopy. Perfusion with GSL-IB4 conjugated to Dylight-594 and fluorescein isothiocyanate was performed to compare against labeling with TL. Co-labeling of choroidal endothelial cells with perfused TL on cross-sections with antibodies directed against the choriocapillaris-restricted endothelial cell marker CA4 was performed. The percentage of perfused choroidal and retinal vessels was assessed semiquantitatively. One mouse was subjected to thermal laser damage before perfusion to cause retinal and choroidal vasculature ablation. Results Intravascular injection of TL led to consistent, robust labeling of retinal and choroidal vascular walls. On cross-sections, choriocapillaris was co-labeled with CA4 and TL. On flat mount, TL perfusion resulted in better labeling of choroidal vessels using tail/jugular vein injection compared with cardiac perfusion (P < .01). More consistent labeling of the choroidal and retinal vascular trees was observed with TL than with GSL-IB4. Vascular damage caused by laser ablation was detected readily using this method. Conclusions TL injection intravascularly can reliably label normal and ablated choroid and retinal vasculature in mouse in a quick, simple manner. Translational Relevance These data will help to facilitate modeling in rodents for diseases such as age-related macular degeneration, diabetes, and other ischemic/angiogenic processes that can also be used for treatment evaluation.
Collapse
Affiliation(s)
- Chunhua Jiao
- Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals & Clinics, Iowa City, IA, USA.,Institute for Vision Research, Iowa City, IA, USA
| | - Kelsey Adler
- Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Xiuying Liu
- Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals & Clinics, Iowa City, IA, USA.,Institute for Vision Research, Iowa City, IA, USA
| | - Weize Sun
- Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals & Clinics, Iowa City, IA, USA.,Institute for Vision Research, Iowa City, IA, USA
| | - Robert F Mullins
- Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals & Clinics, Iowa City, IA, USA.,Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Elliott H Sohn
- Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals & Clinics, Iowa City, IA, USA.,Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
20
|
Clément-Colmou K, Potiron V, Pietri M, Guillonneau M, Jouglar E, Chiavassa S, Delpon G, Paris F, Supiot S. Influence of Radiotherapy Fractionation Schedule on the Tumor Vascular Microenvironment in Prostate and Lung Cancer Models. Cancers (Basel) 2020; 12:E121. [PMID: 31906502 PMCID: PMC7017121 DOI: 10.3390/cancers12010121] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/13/2019] [Accepted: 12/27/2019] [Indexed: 12/29/2022] Open
Abstract
Background. The tumor vasculature acts as an interface for the primary tumor. It regulates oxygenation, nutrient delivery, and treatment efficacy including radiotherapy. The response of the tumor vasculature to different radiation doses has been disparately reported. Whereas high single doses can induce endothelial cell death, improved vascular functionality has also been described in a various dose range, and few attempts have been made to reconcile these findings. Therefore, we aimed at comparing the effects of different radiation fractionation regimens on the tumor vascular microenvironment. METHODS Lewis lung and prostate PC3 carcinoma-derived tumors were irradiated with regimens of 10 × 2 Gy, 6 × 4 Gy, 3 × 8 Gy or 2 × 12 Gy fractions. The tumor vasculature phenotype and function was evaluated by immunohistochemistry for endothelial cells (CD31), pericytes (desmin, α-SMA), hypoxia (pimonidazole) and perfusion (Hoechst 33342). RESULTS Radiotherapy increased vascular coverage similarly in all fractionation regimens in both models. Vessel density appeared unaffected. In PC3 tumors, hypoxia was decreased and perfusion was enhanced in proportion with the dose per fraction. In LLC tumors, no functional changes were observed at t = 15 days, but increased perfusion was noticed earlier (t = 9-11 days). CONCLUSION The vascular microenvironment response of prostate and lung cancers to radiotherapy consists of both tumor/dose-independent vascular maturation and tumor-dependent functional parameters.
Collapse
Affiliation(s)
- Karen Clément-Colmou
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
- Service de Radiothérapie, Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Vincent Potiron
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Manon Pietri
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Maëva Guillonneau
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Emmanuel Jouglar
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Sophie Chiavassa
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
- Service de Physique Médicale, Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Grégory Delpon
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
- Service de Physique Médicale, Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - François Paris
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| | - Stéphane Supiot
- Centre de Recherche en Cancérologie Immunologie Nantes Angers (CRCINA), Institut National de Santé et de la Recherche Médicale (INSERM) UMR U1232, Centre National de la Recherche Scientifique (CNRS) ERL 6001, Université de Nantes, 44007 Nantes, France; (K.C.-C.); (V.P.); (M.P.); (M.G.); (E.J.); (S.C.); (G.D.); (F.P.)
- Laboratoire de Biologie des Cancers et de Théranostic (LabCT), Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
- Service de Radiothérapie, Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France
| |
Collapse
|
21
|
Takino JI, Sato T, Nagamine K, Hori T. The inhibition of Bax activation-induced apoptosis by RasGRP2 via R-Ras-PI3K-Akt signaling pathway in the endothelial cells. Sci Rep 2019; 9:16717. [PMID: 31723205 PMCID: PMC6854084 DOI: 10.1038/s41598-019-53419-4] [Citation(s) in RCA: 30] [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: 06/27/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Apoptosis of endothelial cells is a very important event in various diseases and angiogenesis. We recently reported that ras guanyl nucleotide releasing protein 2 (RasGRP2), which is a guanine nucleotide exchange factor, was expressed in the human umbilical vein endothelial cells (HUVECs) and that Rap1 activation by its overexpression inhibited apoptosis by suppressing tumor necrosis factor-α induced-reactive oxygen species (ROS) production. However, other signaling pathways and roles of RasGRP2 not mediated via Rap1 are not well understood. Therefore, we compared the Mock (M) and the RasGRP2-stable overexpression (R) immortalized HUVECs using BAM7 and anisomycin, which are apoptosis inducers. BAM7 and anisomycin induced apoptosis without causing ROS production, and such apoptosis was significantly increased in M cells, but not in R cells. RasGRP2 suppressed BAM7- and anisomycin-induced apoptosis, but not via the Rap1 pathway as observed using Rap1 knockdown. Furthermore, RasGRP2 activated not only Rap1 but also R-Ras, and suppressed apoptosis by activating R-Ras-phosphoinositide 3-kinase (PI3K)-Akt signaling pathway. The phosphorylation of Akt by RasGRP2 inhibited Bax translocation by promoting translocation of hexokinase-2 (HK-2) from cytoplasm to mitochondria. Taken together, it was suggested that RasGRP2 suppresses the Bax activation-induced apoptosis by promoting HK-2 translocation to mitochondria via R-Ras-PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Jun-Ichi Takino
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan.
| | - Takuma Sato
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan
| | - Kentaro Nagamine
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan
- Department of Clinical Nutrition, Hiroshima International University, Hiroshima, Japan
| | - Takamitsu Hori
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan
| |
Collapse
|
22
|
CCN1-Yes-Associated Protein Feedback Loop Regulates Physiological and Pathological Angiogenesis. Mol Cell Biol 2019; 39:MCB.00107-19. [PMID: 31262999 DOI: 10.1128/mcb.00107-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/23/2019] [Indexed: 01/08/2023] Open
Abstract
Cellular communication network factor 1 (CCN1) is a dynamically expressed, matricellular protein required for vascular development and tissue repair. The CCN1 gene is a presumed target of Yes-associated protein (YAP), a transcriptional coactivator that regulates cell growth and organ size. Herein, we demonstrate that the CCN1 promoter is indeed a direct genomic target of YAP in endothelial cells (ECs) of new blood vessel sprouts and that YAP deficiency in mice downregulates CCN1 and alters cytoskeletal and mitogenic gene expression. Interestingly, CCN1 overexpression in cultured ECs inactivates YAP in a negative feedback and causes its nuclear exclusion. Accordingly, EC-specific deletion of the CCN1 gene in mice mimics a YAP gain-of-function phenotype, characterized by EC hyperproliferation and blood vessel enlargement. CCN1 brings about its effect by providing cells with a soft compliant matrix that creates YAP-repressive cytoskeletal states. Concordantly, pharmacological inhibition of cell stiffness recapitulates the CCN1 deletion vascular phenotype. Furthermore, adeno-associated virus-mediated expression of CCN1 reversed the pathology of YAP hyperactivation and the subsequent aberrant growth of blood vessels in mice with ischemic retinopathy. Our studies unravel a new paradigm of functional interaction between CCN1 and YAP and underscore the significance of their interplay in the pathogenesis of neovascular diseases.
Collapse
|
23
|
Duan LJ, Fong GH. Developmental vascular pruning in neonatal mouse retinas is programmed by the astrocytic oxygen-sensing mechanism. Development 2019; 146:dev.175117. [PMID: 30910827 PMCID: PMC6503987 DOI: 10.1242/dev.175117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/18/2019] [Indexed: 12/17/2022]
Abstract
Vascular pruning is crucial for normal development, but its underlying mechanisms are poorly understood. Here, we report that retinal vascular pruning is controlled by the oxygen-sensing mechanism in local astrocytes. Oxygen sensing is mediated by prolyl hydroxylase domain proteins (PHDs), which use O2 as a substrate to hydroxylate specific prolyl residues on hypoxia inducible factor (HIF)-α proteins, labeling them for polyubiquitylation and proteasomal degradation. In neonatal mice, astrocytic PHD2 deficiency led to elevated HIF-2α protein levels, expanded retinal astrocyte population and defective vascular pruning. Although astrocytic VEGF-A was also increased, anti-VEGF failed to rescue vascular pruning. However, stimulation of retinal astrocytic growth by intravitreal delivery of PDGF-A was sufficient to block retinal vascular pruning in wild-type mice. We propose that in normal development, oxygen from nascent retinal vasculature triggers PHD2-dependent HIF-2α degradation in nearby astrocytic precursors, thus limiting their further growth by driving them to differentiate into non-proliferative mature astrocytes. The physiological limit of retinal capillary density may be set by astrocytes available to support their survival, with excess capillaries destined for regression.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Li-Juan Duan
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, USA
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, USA .,Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, USA
| |
Collapse
|
24
|
Andrée B, Ichanti H, Kalies S, Heisterkamp A, Strauß S, Vogt PM, Haverich A, Hilfiker A. Formation of three-dimensional tubular endothelial cell networks under defined serum-free cell culture conditions in human collagen hydrogels. Sci Rep 2019; 9:5437. [PMID: 30932006 PMCID: PMC6443732 DOI: 10.1038/s41598-019-41985-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/20/2019] [Indexed: 12/15/2022] Open
Abstract
Implementation of tubular endothelial cell networks is a prerequisite for 3D tissue engineering of constructs with clinically relevant size as nourishment of cells is challenged by the diffusion limit. In vitro generation of 3D networks is often achieved under conditions using serum containing cell culture medium and/or animal derived matrices. Here, 3D endothelial cell networks were generated by using human umbilical vein endothelial cells (HUVECs) in combination with human adipose tissue derived stromal cells (hASCs) employing human collagen I as hydrogel and decellularized porcine small intestinal submucosa as starter matrix. Matrigel/rat tail collagen I hydrogel was used as control. Resulting constructs were cultivated either in serum-free medium or in endothelial growth medium-2 serving as control. Endothelial cell networks were quantified, tested for lumen formation, and interaction of HUVECs and hASCs. Tube diameter was slightly larger in constructs containing human collagen I compared to Matrigel/rat tail collagen I constructs under serum-free conditions. All other network parameters were mostly similar. Thereby, the feasibility of generating 3D endothelial cell networks under serum-free culture conditions in human collagen I as hydrogel was demonstrated. In summary, the presented achievements pave the way for the generation of clinical applicable constructs.
Collapse
Affiliation(s)
- Birgit Andrée
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Houda Ichanti
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Stefan Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Sarah Strauß
- Department of Plastic, Asthetic, Hand- and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Peter-Maria Vogt
- Department of Plastic, Asthetic, Hand- and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
25
|
Mazio C, Casale C, Imparato G, Urciuolo F, Attanasio C, De Gregorio M, Rescigno F, Netti PA. Pre-vascularized dermis model for fast and functional anastomosis with host vasculature. Biomaterials 2019; 192:159-170. [DOI: 10.1016/j.biomaterials.2018.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 12/16/2022]
|
26
|
Lara E, Acurio J, Leon J, Penny J, Torres-Vergara P, Escudero C. Are the Cognitive Alterations Present in Children Born From Preeclamptic Pregnancies the Result of Impaired Angiogenesis? Focus on the Potential Role of the VEGF Family. Front Physiol 2018; 9:1591. [PMID: 30487752 PMCID: PMC6246680 DOI: 10.3389/fphys.2018.01591] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022] Open
Abstract
Evidence from clinical studies has proposed that children born from preeclamptic women have a higher risk of suffering neurological, psychological, or behavioral alterations. However, to date, the mechanisms behind these outcomes are poorly understood. Here, we speculate that the neurodevelopmental alterations in the children of preeclamptic pregnancies result from impaired angiogenesis. The pro-angiogenic factors vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) are key regulators of both vascular and neurological development, and it has been widely demonstrated that umbilical blood of preeclamptic pregnancies contains high levels of soluble VEGF receptor type 1 (sFlt-1), a decoy receptor of VEGF. As a consequence, this anti-angiogenic state could lead to long-lasting neurological outcomes. In this non-systematic review, we propose that alterations in the circulating concentrations of VEGF, PlGF, and sFlt-1 in preeclamptic pregnancies will affect both fetal cerebrovascular function and neurodevelopment, which in turn may cause cognitive alterations in post-natal life.
Collapse
Affiliation(s)
- Evelyn Lara
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - José Leon
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Department of Pharmacy, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Red Iberoamericana de alteraciones Vasculares Asociadas a TRastornos del EMbarazo (RIVA-TREM), Chillán, Chile
| |
Collapse
|
27
|
Hao W, Han J, Chu Y, Huang L, Zhuang Y, Sun J, Li X, Zhao Y, Chen Y, Dai J. Collagen/Heparin Bi‐Affinity Multilayer Modified Collagen Scaffolds for Controlled bFGF Release to Improve Angiogenesis In Vivo. Macromol Biosci 2018; 18:e1800086. [DOI: 10.1002/mabi.201800086] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/07/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Wangping Hao
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Jie Han
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
- Xi’an Jiaotong University Xi’an 710049 P.R. China
| | - Yun Chu
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Lei Huang
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Yan Zhuang
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Jie Sun
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Xiaoran Li
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Yannan Zhao
- Xi’an Jiaotong University Xi’an 710049 P.R. China
- Center for Regenerative MedicineState Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of Sciences Beijing 100101 P.R. China
| | - Yanyan Chen
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
| | - Jianwu Dai
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of Sciences Suzhou 215123 P.R. China
- Xi’an Jiaotong University Xi’an 710049 P.R. China
- Center for Regenerative MedicineState Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of Sciences Beijing 100101 P.R. China
| |
Collapse
|
28
|
Nagaraju S, Truong D, Mouneimne G, Nikkhah M. Microfluidic Tumor-Vascular Model to Study Breast Cancer Cell Invasion and Intravasation. Adv Healthc Mater 2018; 7:e1701257. [PMID: 29334196 DOI: 10.1002/adhm.201701257] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/14/2017] [Indexed: 12/16/2022]
Abstract
Cancer is a major leading cause of disease-related death in the world. The severe impact of cancer can be attributed to poor understanding of the mechanisms involved in earliest steps of the metastatic cascade, specifically invasion into the surrounding stroma and intravasation into the blood capillaries. However, conducting integrated biological studies of invasion and intravasation have been challenging, within in vivo models and traditional in vitro assay, due to difficulties in establishing a precise tumor microenvironment. To that end, in this work, a novel 3D microfluidic platform comprised of concentric three-layer cell-laden hydrogels for simultaneous investigation of breast cancer cell invasion and intravasation as well as vasculature maturation influenced by tumor-vascular crosstalk is developed. It was demonstrated that the presence of spontaneously formed vasculature enhance MDA-MB-231 invasion into the 3D stroma. Following invasion, cancer cells are visualized intravasating into the outer vasculature. Additionally, invading cancer cells significantly reduce vessel diameter while increasing permeability, consistent with previous in vivo studies. Major signaling cytokines involved in tumor-vascular crosstalk that govern cancer cell invasion and intravasation are further identified. Taken together, this platform will enable unique insights of critical biological events within the metastatic cascade, with significant potential for developing efficient cancer therapeutics.
Collapse
Affiliation(s)
- Supriya Nagaraju
- School of Biological and Health Systems Engineering (SBHSE) Arizona State University Tempe AZ 85287 USA
| | - Danh Truong
- School of Biological and Health Systems Engineering (SBHSE) Arizona State University Tempe AZ 85287 USA
| | | | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE) Arizona State University Tempe AZ 85287 USA
| |
Collapse
|
29
|
Neto F, Klaus-Bergmann A, Ong YT, Alt S, Vion AC, Szymborska A, Carvalho JR, Hollfinger I, Bartels-Klein E, Franco CA, Potente M, Gerhardt H. YAP and TAZ regulate adherens junction dynamics and endothelial cell distribution during vascular development. eLife 2018; 7:31037. [PMID: 29400648 PMCID: PMC5814147 DOI: 10.7554/elife.31037] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/02/2018] [Indexed: 12/15/2022] Open
Abstract
Formation of blood vessel networks by sprouting angiogenesis is critical for tissue growth, homeostasis and regeneration. How endothelial cells arise in adequate numbers and arrange suitably to shape functional vascular networks is poorly understood. Here we show that YAP/TAZ promote stretch-induced proliferation and rearrangements of endothelial cells whilst preventing bleeding in developing vessels. Mechanistically, YAP/TAZ increase the turnover of VE-Cadherin and the formation of junction associated intermediate lamellipodia, promoting both cell migration and barrier function maintenance. This is achieved in part by lowering BMP signalling. Consequently, the loss of YAP/TAZ in the mouse leads to stunted sprouting with local aggregation as well as scarcity of endothelial cells, branching irregularities and junction defects. Forced nuclear activity of TAZ instead drives hypersprouting and vascular hyperplasia. We propose a new model in which YAP/TAZ integrate mechanical signals with BMP signaling to maintain junctional compliance and integrity whilst balancing endothelial cell rearrangements in angiogenic vessels.
Collapse
Affiliation(s)
- Filipa Neto
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Vascular Biology Laboratory, Lincoln's Inn Fields Laboratories, London Research Institute - Cancer Research UK, London, United Kingdom
| | - Alexandra Klaus-Bergmann
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Yu Ting Ong
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Silvanus Alt
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Anne-Clémence Vion
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Anna Szymborska
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Joana R Carvalho
- Vascular Morphogenesis Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | | | - Eireen Bartels-Klein
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Claudio A Franco
- Vascular Morphogenesis Laboratory, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Michael Potente
- DZHK (German Center for Cardiovascular Research), Berlin, Germany.,Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Holger Gerhardt
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Vascular Biology Laboratory, Lincoln's Inn Fields Laboratories, London Research Institute - Cancer Research UK, London, United Kingdom.,DZHK (German Center for Cardiovascular Research), Berlin, Germany.,Vascular Patterning Laboratory, Vesalius Research Center, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium.,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
30
|
Watson EC, Grant ZL, Coultas L. Endothelial cell apoptosis in angiogenesis and vessel regression. Cell Mol Life Sci 2017; 74:4387-4403. [PMID: 28646366 PMCID: PMC11107683 DOI: 10.1007/s00018-017-2577-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Blood vessel regression is an essential process for ensuring blood vessel networks function at optimal efficiency and for matching blood supply to the metabolic needs of tissues as they change over time. Angiogenesis is the major mechanism by which new blood vessels are produced, but the vessel growth associated with angiogenesis must be complemented by remodeling and maturation events including the removal of redundant vessel segments and cells to fashion the newly forming vasculature into an efficient, hierarchical network. This review will summarize recent findings on the role that endothelial cell apoptosis plays in vascular remodeling during angiogenesis and in vessel regression more generally.
Collapse
Affiliation(s)
- Emma C Watson
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
- Faculty of Medicine, University of Münster, 48149, Münster, Germany
| | - Zoe L Grant
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Leigh Coultas
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
31
|
Wu S, Peng H, Li X, Streubel PN, Liu Y, Duan B. Effect of scaffold morphology and cell co-culture on tenogenic differentiation of HADMSC on centrifugal melt electrospun poly (L‑lactic acid) fibrous meshes. Biofabrication 2017; 9:044106. [PMID: 29134948 PMCID: PMC5849472 DOI: 10.1088/1758-5090/aa8fb8] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Engineered tendon grafts offer a promising alternative for grafting during the reconstruction of complex tendon tears. The tissue-engineered tendon substitutes have the advantage of increased biosafety and the option to customize their biochemical and biophysical properties to promote tendon regeneration. In this study, we developed a novel centrifugal melt electrospinning (CME) technique, with the goal of optimizing the fabrication parameters to generate fibrous scaffolds for tendon tissue engineering. The effects of CME processing parameters, including rotational speed, voltage, and temperature, on fiber properties (i.e. orientation, mean diameter, and productivity) were systematically investigated. By using this solvent-free and environmentally friendly method, we fabricated both random and aligned poly (L-lactic acid) (PLLA) fibrous scaffolds with controllable mesh thickness. We also investigated and compared their morphology, surface hydrophilicity, and mechanical properties. We seeded human adipose derived mesenchymal stem cells (HADMSC) on various PLLA fibrous scaffolds and conditioned the constructs in tenogenic differentiation medium for up to 21 days, to investigate the effects of fiber alignment and scaffold thickness on cell behavior. Aligned fibrous scaffolds induced cell elongation and orientation through a contact guidance phenomenon and promoted HADMSC proliferation and differentiation towards tenocytes. At the early stage, thinner scaffolds were beneficial for HADMSC proliferation, but the scaffold thickness had no significant effects on cell proliferation for longer-term cell culture. We further co-seeded HADMSC and human umbilical vein endothelial cells (HUVEC) on aligned PLLA fibrous mats and determined how the vascularization affected HADMSC tenogenesis. We found that co-cultured HADMSC-HUVEC expressed more tendon-related markers on the aligned fibrous scaffold. The co-culture systems promoted in vitro HADMSC differentiation towards tenocytes. These aligned fibrous scaffolds fabricated by CME technique could potentially be utilized to repair and regenerate tendon defects and injuries with cell co-culture and controlled vascularization.
Collapse
Affiliation(s)
- Shaohua Wu
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hao Peng
- College of Mechanical and Electric Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiuhong Li
- College of Mechanical and Electric Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Philipp N. Streubel
- Department of Orthopedic Surgery and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yong Liu
- College of Mechanical and Electric Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
32
|
Retinal vasculature development in health and disease. Prog Retin Eye Res 2017; 63:1-19. [PMID: 29129724 DOI: 10.1016/j.preteyeres.2017.11.001] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/17/2022]
Abstract
Development of the retinal vasculature is based on highly coordinated signalling between different cell types of the retina, integrating internal metabolic requirements with external influences such as the supply of oxygen and nutrients. The developing mouse retinal vasculature is a useful model system to study these interactions because it is experimentally accessible for intra ocular injections and genetic manipulations, can be easily imaged and develops in a similar fashion to that of humans. Research using this model has provided insights about general principles of angiogenesis as well as pathologies that affect the developing retinal vasculature. In this review, we discuss recent advances in our understanding of the molecular and cellular mechanisms that govern the interactions between neurons, glial and vascular cells in the developing retina. This includes a review of mechanisms that shape the retinal vasculature, such as sprouting angiogenesis, vascular network remodelling and vessel maturation. We also explore how the disruption of these processes in mice can lead to pathology - such as oxygen induced retinopathy - and how this translates to human retinopathy of prematurity.
Collapse
|
33
|
Vascular heterogeneity and specialization in development and disease. Nat Rev Mol Cell Biol 2017; 18:477-494. [PMID: 28537573 DOI: 10.1038/nrm.2017.36] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Blood and lymphatic vessels pervade almost all body tissues and have numerous essential roles in physiology and disease. The inner lining of these networks is formed by a single layer of endothelial cells, which is specialized according to the needs of the tissue that it supplies. Whereas the general mechanisms of blood and lymphatic vessel development are being defined with increasing molecular precision, studies of the processes of endothelial specialization remain mostly descriptive. Recent insights from genetic animal models illuminate how endothelial cells interact with each other and with their tissue environment, providing paradigms for vessel type- and organ-specific endothelial differentiation. Delineating these governing principles will be crucial for understanding how tissues develop and maintain, and how their function becomes abnormal in disease.
Collapse
|
34
|
Lateral thinking - Interocular symmetry and asymmetry in neurovascular patterning, in health and disease. Prog Retin Eye Res 2017; 59:131-157. [PMID: 28457789 DOI: 10.1016/j.preteyeres.2017.04.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/24/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
Abstract
No biological system or structure is likely to be perfectly symmetrical, or have identical right and left forms. This review explores the evidence for eye and visual pathway asymmetry, in health and in disease, and attempts to provide guidance for those studying the structure and function of the visual system, where recognition of symmetry or asymmetry may be essential. The principal question with regards to asymmetry is not 'are the eyes the same?', for some degree of asymmetry is pervasive, but 'when are they importantly different?'. Knowing if right and left eyes are 'importantly different' could have significant consequences for deciding whether right or left eyes are included in an analysis or for examining the association between a phenotype and ocular parameter. The presence of significant asymmetry would also have important implications for the design of normative databases of retinal and optic nerve metrics. In this review, we highlight not only the universal presence of asymmetry, but provide evidence that some elements of the visual system are inherently more asymmetric than others, pointing to the need for improved normative data to explain sources of asymmetry and their impact on determining associations with genetic, environmental or health-related factors and ultimately in clinical practice.
Collapse
|
35
|
Barbacena P, Carvalho JR, Franco CA. Endothelial cell dynamics in vascular remodelling. Clin Hemorheol Microcirc 2017; 64:557-563. [PMID: 27802214 DOI: 10.3233/ch-168006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this ESCHM 2016 conference talk report, we summarise two recently published original articles Franco et al. PLoS Biology 2015 and Franco et al. eLIFE 2016. The vascular network undergoes extensive vessel remodelling to become fully functional. Is it well established that blood flow is a main driver for vascular remodelling. It has also been proposed that vessel pruning is a central process within physiological vessel remodelling. However, despite its central function, the cellular and molecular mechanisms regulating vessel regression, and their interaction with blood flow patterns, remain largely unexplained. We investigated the cellular process governing developmental vascular remodelling in mouse and zebrafish. We established that polarised reorganization of endothelial cells is at the core of vessel regression, representing vessel anastomosis in reverse. Moreover, we established for the first time an axial polarity map for all endothelial cells together with an in silico method for the computation of the haemodynamic forces in the murine retinal vasculature. Using network-level analysis and microfluidics, we showed that endothelial non-canonical Wnt signalling regulates endothelial sensitivity to shear forces. Loss of Wnt5a/11 renders endothelial cells more sensitive to shear, resulting in axial polarisation at lower shear stress levels. Collectively our data suggest that non-canonical Wnt signalling stabilizes forming vascular networks by reducing endothelial shear sensitivity, thus keeping vessels open under low flow conditions that prevail in the primitive plexus.
Collapse
|